EP2121620B1 - Inhibitors of mek - Google Patents

Inhibitors of mek Download PDF

Info

Publication number
EP2121620B1
EP2121620B1 EP08727965.9A EP08727965A EP2121620B1 EP 2121620 B1 EP2121620 B1 EP 2121620B1 EP 08727965 A EP08727965 A EP 08727965A EP 2121620 B1 EP2121620 B1 EP 2121620B1
Authority
EP
European Patent Office
Prior art keywords
fluoro
methyl
compound
mmol
solution
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
EP08727965.9A
Other languages
German (de)
French (fr)
Other versions
EP2121620A1 (en
Inventor
Jean-Michel Vernier
Andreas Maderna
Yung-Hyo Koh
Zhi Hong
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Ardea Biociences Inc
Original Assignee
Ardea Biociences Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Ardea Biociences Inc filed Critical Ardea Biociences Inc
Publication of EP2121620A1 publication Critical patent/EP2121620A1/en
Application granted granted Critical
Publication of EP2121620B1 publication Critical patent/EP2121620B1/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D231/00Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings
    • C07D231/54Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings condensed with carbocyclic rings or ring systems
    • C07D231/56Benzopyrazoles; Hydrogenated benzopyrazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4151,2-Diazoles
    • A61K31/4161,2-Diazoles condensed with carbocyclic ring systems, e.g. indazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D235/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings
    • C07D235/02Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings condensed with carbocyclic rings or ring systems
    • C07D235/04Benzimidazoles; Hydrogenated benzimidazoles
    • C07D235/06Benzimidazoles; Hydrogenated benzimidazoles with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached in position 2
    • C07D235/14Radicals substituted by nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/70Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings condensed with carbocyclic rings or ring systems
    • C07D239/72Quinazolines; Hydrogenated quinazolines
    • C07D239/74Quinazolines; Hydrogenated quinazolines with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, attached to ring carbon atoms of the hetero ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D261/00Heterocyclic compounds containing 1,2-oxazole or hydrogenated 1,2-oxazole rings
    • C07D261/20Heterocyclic compounds containing 1,2-oxazole or hydrogenated 1,2-oxazole rings condensed with carbocyclic rings or ring systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems

Definitions

  • Ras/Raf kinase pathway is one of the most important and most well understood MAP kinase pathways involved in normal and uncontrolled cell growth.
  • Active GTP-bound Ras leads to the activation of a cascade of serine/threonine kinases.
  • One of the several groups of kinases known to require GTP-bound Ras for their activation is the Raf family. Upon activation Raf phosphorylates "mitogen-activated ERK activating kinases" (MEK) - 1 and MEK2.
  • Some cancers contain mutations which result in the continuous activation of this pathway due to continuous production of growth factors. Other mutations can lead to defects in the deactivation of the activated GTP-bound Ras complex, again resulting in activation of the MAP kinase pathway. Mutated, oncogenic forms of Ras are found in 50% of colon and >90% pancreatic cancers as well as many others types of cancers. Recently, bRaf mutations have been identified in more than 60% of malignant melanoma.
  • MEK is a key player in this pathway as it is downstream of Ras and Raf. Additionally, it is an attractive therapeutic target because the only known substrates for MEK phosphorylation are the MAP kinases, ERK1 and 2.
  • MEK inhibitors have been shown to inhibit human tumor growth in nude mouse xenografts, block static allodynia in animals and inhibit growth of acute myeloid leukemia cells.
  • MEK1 and MEK2 are validated and accepted targets for anti-proliferative therapies, even when the oncogenic mutation does not affect MEK structure or expression.
  • the MEK cascade has also been implicated in inflammatory diseases and disorders. This includes both acute and chronic inflammation disorders. Examples of such disorders are allergic contact dermatitis, rheumatoid arthritis, osteoarthritis, inflammatory bowel diseases, chronic obstructive pulmonary disorder, psoriasis, multiple sclerosis, asthma, diseases and disorders related to diabetic complications, and inflammatory complications of the cardiovascular system such as acute coronary syndrome. Among inflammatory bowel diseases are Crohn's disease and ulcerative colitis.
  • European patent application published under EP 0 679 641 A (Eisai Co Ltd) relates to sulfonamide derivatives and sulfonic ester derivatives having anti-tumour activity.
  • the invention provides compounds and methods of utilizing such compounds.
  • the compounds and the pharmaceutically acceptable salts, esters, hydrates and solvates thereof of this invention are useful, e.g. in the treatment of diseases, e.g. hyperproliferative diseases.
  • the invention provides compounds, including pharmaceutically acceptable salts, esters, hydrates and solvates thereof having the general formula I: where G is R 1a , R 1b , R 1c , R 1d , R 1e , Ar 1 , Ar 2 or Ar 3 ; R o , R 1 and R 2 are independently selected from H, halogen, cyano, cyanomethyl, nitro, difluoromethoxy, difluoromethoxy, trifluoromethyl, azido, CO 2 R 5 , OR 5 , -O-(CO)-R 5 , -O-C(O)-N(R 5 ) 2 , -NR 5 C(O)NR 6 R 7 , -SR 5 , NHC(O
  • R 1a is methyl, optionally substituted with 1-3 fluorine atoms or 1-3 chlorine atoms, or with OH, cyclopropoxy, or C1- C4 alkoxy, where the C1- C4 alkyl moieties of said C1- C4 alkoxy groups are optionally substituted with one hydroxy or methoxy group, and where all C2- C4 alkyl groups within said C1- C4 alkoxy are optionally further substituted with a second OH group.
  • R 1b is CH(CH 3 )-C1-3 alkyl or C3-C6 cycloalkyl, said methyl, alkyl, and cycloalkyl groups optionally substituted with 1-3 substituents selected independently from F, Cl, Br, I, OH, C1-C4 alkoxy, and CN.
  • R 1c is (CH 2 ) n O m R', where m is 0 or 1; where, when m is 1, n is 2 or 3, and when m is 0, n is 1 or 2; and where R' is C1-C6 alkyl, optionally substituted with 1-3 substituents selected independently from F, Cl, OH, OCH 3 , OCH 2 CH 3 , and C3-C6 cycloalkyl;
  • R 1d is C(A')(A")(B)- where B, A', and A" are, independently, H or C1-4 alkyl, optionally substituted with one or two OH groups or halogen atoms, or A' and A", together with the carbon atom to which they are attached, form a 3- to 6- member saturated ring, said ring optionally containing one or two heteroatoms selected, independently, from O, N, and S and optionally substituted with one or two groups selected independently from methyl, ethyl, and halo.
  • R 1e is benzyl or 2-phenyl ethyl, in which the phenyl group is optionally substituted where q is 1 or 2,
  • Rs, R 9 and R 10 are, independently, H, F, Cl, Br, I, CH 3 , CH 2 F, CHF 2 , CF 3 , OCH 3 , OCH 2 F, OCHF 2 , OCF 3 , ethyl, n -propyl, isopropyl, cyclopropyl, isobutyl, sec -butyl, tert -butyl, and methylsulfonyl
  • R 10 may also be nitro, acetamido, amidinyl, cyano, carbamoyl, methylcarbamoyl, dimethylcarbamoyl, 1,3,4-oxadiazol-2-yl, 5-methyl-1,3,4-5 oxadiazolyl, 1,3,4-thiadiazolyl, 5-methyl-1,3,4-thi
  • Ar 1 is where W and V are, independently, N, CR 2 or CR 3 ;
  • R 3 , Rs, R 8 and R 10 are, independently, H, F, Cl, Br, I, CH 3 , CH 2 F, CHF 2 , CF 3 , OCH 3 , OCH 2 F, OCHF 2 , OCF 3 , ethyl, n -propyl, isopropyl, cyclopropyl, isobutyl, sec -butyl, tert -butyl, and methylsulfonyl, and R 10 may also be nitro, acetamido, amidinyl, cyano, carbamoyl, methylcarbamoyl, dimethylcarbamoyl, 1,3,4-oxadiazol-2-yl, 5-methyl-1,3,4-oxadiazol, 1,3,4-thiadiazol, 5-methyl-1,3,4-thiadiazol 1H-tetrazolyl, N-morpholiny
  • Ar 3 is where W is -NH-, -NCH3- or -O-; and R 13 and R 8 are, independently, H, F, Cl, or methyl.
  • Such compounds are inhibitors of MEK and are useful in treatment of cancer and other hyperproliferative diseases.
  • compositions comprising pharmaceutically effective amounts of a compound of formula I or a pharmaceutically acceptable salt, ester, solvate or hydrate thereof, and a pharmaceutically acceptable carrier.
  • Such compositions may contain adjuvants, excipients, preservatives, agents for delaying absorption, fillers, binders, adsorbents, buffers, disintegrating agents, solubilizing agents, other carriers, and other inert ingredients. Methods of formulation of such compositions are well-known in the art.
  • the invention is also directed to use of a compound of formula I, or a pharmaceutically acceptable salt, ester, solvate or hydrate thereof, for the preparation of a medicament for the treatment of a hyperproliferative disease.
  • the invention is also directed to use of a compound of formula I, or a pharmaceutically acceptable salt, ester, solvate or hydrate thereof, for the preparation of a medicament for the treatment of a disorder or condition modulated by the MEK cascade.
  • the appropriate dosage for a particular patient can be determined, according to known methods, by those skilled in the art.
  • Novel compounds encompassed by the instant invention include those described by the general formulas I and II set forth above, and the pharmaceutically acceptable salts, esters,solvates or hydrates thereof.
  • a "pharmaceutically acceptable salt” includes salts that retain the biological effectiveness of the free acids and bases of the specified compound and that are not biologically or otherwise undesirable.
  • a compound of this invention may possess acidic or basic groups and therefore may react with any of a number of inorganic or organic bases, and inorganic and organic acids, to form a pharmaceutically acceptable salt.
  • Examples of pharmaceutically acceptable salts include those salts prepared by reaction of the compounds of this invention with a mineral or organic acid or an inorganic base, such salts including sulfates, pyrosulfates, bisulfates, sulfites, bisulfites, phosphates, monohydrogenphosphates, dihydrogenphosphates, metaphosphates, pyrophosphates, chlorides, bromides, iodides, acetates, propionates, decanoates, caprylates, acrylates, formates, isobutyrates, caproates, heptanoates, propiolates, oxalates, malonates, succinates, suberates, sebacates, fumarates, maleates, butyn-1,4-dioates, hexyne-1,6-dioates, benzoates, chlorobenzoates, methylbenzoates, dinitrobenzoates, hydroxybenzoates, methoxybenzoates, phthal
  • the invention provides compounds, including pharmaceutically acceptable saltsand solvates thereof having the formula : wherein
  • the present invention provides compounds of formula (I-A), (I-B), (I-C) or (I-D), where X is F, Cl, or CH 3 ; Y is I, Br, Cl, CF 3 , or C1-C3 alkyl; and Z is H or F.
  • the present invention provides compounds of formula (I-A), (I-B), (I-C) or (I-D), where R o is F, Cl, C1-C4 alkyl or C1-C4 alkoxy, said C1-C4 alkyl group and the C1-C4 alkyl moiety of said C1-C4 alkoxy group optionally substituted with F, Cl, OCH 3 , or OCH 2 CH 3 .
  • the present invention provides compounds of formula (I-A), (I-B), (I-C) or (I-D), where R o is H, F, Cl, C1-C4 alkyl, methoxy, ethoxy, or 2-methoxy-ethoxy.
  • the present invention provides compounds of formula (I-A), (I-B9, (I-C) or (I-D), where G is R 1d .
  • the present invention provides compounds of formula (I-A), (I-B), (I-C) or (I-D), where R o is either :
  • the present invention provides compounds of formula (I-A), (I-B), (I-C) or (I-D), where C(A')(A") is C1-C6 cycloalkyl.
  • the present invention provides compounds of formula (I-A), (I-B), (I-C) or (I-D), where B is H.
  • the present invention provides compounds of formula (I-A), (I-B), (I-C) or (I-D), where C(A')(A") is cyclopropyl.
  • the present invention provides compounds of formula (I-A), (I-B), (I-C) or (I-D), where B is methyl, optionally substituted with one OH group, or C2-C4 alkyl, optionally substituted with one or two OH groups.
  • the present invention provides compounds of formula (I-A), (I-B), (I-C) or (I-D), where C(A')(A") is cyclopropyl.
  • the present invention provides compounds of formula (I-A), (I-B), (I-C) or (I-D), where B is methyl, ethyl, 2-hydroxyethyl, n-propyl, 3-5 hydroxypropyl, 2,3-dihydroxypropyl, 3,4-dihydroxybutyl, isopropyl, 1-methyl-2-hydroxyethyl, n-butyl, sec-butyl, isobutyl, or 2-hydroxymethyl-3-hydroxy propyl.
  • the present invention provides compounds of formula (I-A), (I-B), (I-C) or (I-D), where B is 2,3-dihydroxypropyl or 3,4-dihydroxybutyl.
  • the present invention provides compounds of formula (I-A), (I-B), (I-C) or (I-D), in which the chiral carbon in B is in the R configuration.
  • Certain compounds of the present invention may have asymmetric centers and therefore exist in different enantiomeric forms. All optical isomers and stereoisomers of the compounds of the present invention, and mixtures thereof, are considered to be within the scope of the invention.
  • the invention includes the use of a racemate, one or more enantiomeric forms, one or more diastereomeric forms, or mixtures thereof.
  • the compounds of the present invention may also exist as tautomers. This invention relates to the use of all such tautomers and mixtures thereof.
  • the compounds of the present invention may also exist in different polymorphic states. This invention relates to the use of all such polymorphic states and mixtures thereof.
  • the subject invention also includes isotopically-labeled compounds, which are identical to those recited in the present invention, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature.
  • isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, sulfur, fluorine and chloride, such as 2 H, 3 H, 13 C, 14 C, 15 N, 18 0, 17 O, 31 P, 32 P, 35 S, 18 F, and 36 Cl, respectively.
  • Tritiated, i. e., 3 H and carbon-14, i. e., 14 C, isotopes are particularly preferred for their ease of preparation and detectability. Further, substitution with heavier isotopes such as deuterium, i. e., 2 H, can afford certain therapeutic advantages resulting from greater metabolic stability, for example increased in vivo half-life or reduced dosage requirements and, hence, may be preferred in some circumstances.
  • Isotopically labeled compound of the present invention can generally be prepared by carrying out procedures disclosed in the Schemes and/or in the Examples and Preparations below, by substituting a readily available isotopically labeled reagent for a non-isotopically labeled reagent.
  • radical arylalkyl is attached to the structure in question by the alkyl group.
  • the invention also relates to pharmaceutical compositions.
  • the invention provides pharmaceutical compositions for the treatment of disorders such as hyperproliferative disorder in a mammal.
  • the treatment of said disorders comprises a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt, ester, solvate or hydrate thereof, and a pharmaceutically acceptable carrier.
  • said pharmaceutical composition is for the treatment of cancer such as acute myeloid leukemia, thymus, brain, lung, squamous cell, skin, eye, retinoblastoma, intraocular melanoma, oral cavity and oropharyngeal, bladder, gastric, stomach, pancreatic, bladder, breast, cervical, head, neck, renal, kidney, liver, ovarian, prostate, colorectal, esophageal, testicular, gynecological, thyroid, CNS, PNS, AIDS related AIDS-Related (e.g. Lymphoma and Kaposi's Sarcoma) or Viral-Induced cancer.
  • cancer such as acute myeloid leukemia, thymus, brain, lung, squamous cell, skin, eye, retinoblastoma, intraocular melanoma, oral cavity and oropharyngeal, bladder, gastric, stomach, pancreatic, bladder, breast, cervical, head, neck, renal, kidney,
  • said pharmaceutical composition is for the treatment of a non-cancerous hyperproliferative disorder such as benign hyperplasia of the skin (e. g., psoriasis), restenosis, or prostate (e. g., benign prostatic hypertrophy (BPH)).
  • a non-cancerous hyperproliferative disorder such as benign hyperplasia of the skin (e. g., psoriasis), restenosis, or prostate (e. g., benign prostatic hypertrophy (BPH)).
  • the invention also relates to a pharmaceutical composition for the treatment of pancreatitis or kidney disease (including proliferative glomerulonephritis and diabetes- induced renal disease) or pain in a mammal which comprises a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt, ester, solvate or hydrate thereof, and a pharmaceutically acceptable carrier.
  • the invention also relates to a pharmaceutical composition for the prevention of blastocyte implantation in a mammal which comprises a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt, ester, solvate or hydrate thereof, and a pharmaceutically acceptable carrier.
  • the invention also relates to a pharmaceutical composition for treating a disease related to vasculogenesis or angiogenesis in a mammal which comprises a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt, ester, solvate or hydrate thereof, and a pharmaceutically acceptable carrier.
  • said pharmaceutical composition is for treating a disease selected from the group consisting of tumor angiogenesis, chronic inflammatory disease such as rheumatoid arthritis, inflammatory bowel disease, atherosclerosis, skin diseases such as psoriasis, eczema, and scleroderma, diabetes, diabetic retinopathy, retinopathy of prematurity, age-related macular degeneration, hemangioma, glioma, melanoma, Kaposi's sarcoma and ovarian, breast, lung, pancreatic, prostate, colon and epidermoid cancer.
  • a disease selected from the group consisting of tumor angiogenesis, chronic inflammatory disease such as rheumatoid arthritis, inflammatory bowel disease, atherosclerosis, skin diseases such as psoriasis, eczema, and scleroderma
  • diabetes diabetic retinopathy, retinopathy of prematurity, age-related macular degeneration, heman
  • the invention also relates to use of a compound of the present invention, or a pharmaceutically acceptable salt, ester, solvate or hydrate thereof for the preparation of a medicament for treating a hyperproliferative disorder.
  • said use is in the treatment of cancer such as acute myeloid leukemia, thymus, brain, lung, squamous cell, skin, eye, retinoblastoma, intraocular melanoma, oral cavity and oropharyngeal, bladder, gastric, stomach, pancreatic, bladder, breast, cervical, head, neck, renal, kidney, liver, ovarian, prostate, colorectal, esophageal, testicular, gynecological, thyroid, CNS, PNS, AIDS related AIDS-Related (e.g.
  • cancer such as acute myeloid leukemia, thymus, brain, lung, squamous cell, skin, eye, retinoblastoma, intraocular melanoma, oral cavity and or
  • said use is in the treatment of a non-cancerous hyperproliferative disorder such as benign hyperplasia of the skin (e. g., psoriasis), restenosis, or prostate (e. g., benign prostatic hypertrophy (BPH)).
  • a non-cancerous hyperproliferative disorder such as benign hyperplasia of the skin (e. g., psoriasis), restenosis, or prostate (e. g., benign prostatic hypertrophy (BPH)).
  • the invention also relates to use of a compound of the present invention, or a pharmaceutically acceptable salt, ester, solvate or hydrate thereof, in combination with an anti-tumor agent for the preparation of a medicament for the treatment of a hyperproliferative disorder.
  • the anti-tumor agent is selected from the group consisting of mitotic inhibitors, alkylating agents, anti- metabolites, intercalating antibiotics, growth factor inhibitors, cell cycle inhibitors, enzyme inhibitors, topoisomerase inhibitors, biological response modifiers, anti- hormones, angiogenesis inhibitors, and anti-androgens.
  • the invention also relates to use of a compound of the present invention, or a pharmaceutically acceptable salt, ester, solvate or hydrate thereof, for the preparation of a medicament for the treatment of diseases related to vasculogenesis or angiogenesis.
  • said use is in treating a disease selected from the group consisting of tumor angiogenesis, chronic inflammatory disease such as rheumatoid arthritis, atherosclerosis, inflammatory bowel disease, skin diseases such as psoriasis, eczema, and scleroderma, diabetes, diabetic retinopathy, retinopathy of prematurity, age-related macular degeneration, hemangioma, glioma, melanoma, Kaposi's sarcoma and ovarian, breast, lung, pancreatic, prostate, colon and epidermoid cancer.
  • Patients that can be treated with compounds of the present invention, or pharmaceutically acceptable salt, ester, solvate or hydrate of said compounds include, for example, patients that have been diagnosed as having psoriasis; restenosis; atherosclerosis; BPH; breast cancer such as a ductal carcinoma in duct tissue in a mammary gland, medullary carcinomas, colloid carcinomas, tubular carcinomas, and inflammatory breast cancer; ovarian cancer, including epithelial ovarian tumors such as adenocarcinoma in the ovary and an adenocarcinoma that has migrated from the ovary into the abdominal cavity; uterine cancer; cervical cancer such as adenocarcinoma in the cervix epithelial including squamous cell carcinoma and adenocarcinomas; prostate cancer, such as a prostate cancer selected from the following: an adenocarcinoma or an adenocarinoma that has migrated to the bone; pancreatic cancer such as epitheli
  • the invention further provides methods of modulating MEK activity by contacting MEK with an amount of a compound of the invention sufficient to modulate the activity of MEK. Modulate can be inhibiting or activating MEK activity. In some embodiments, the invention provides methods of inhibiting MEK activity by contacting MEK with an amount of a compound of the invention sufficient to inhibit the activity of MEK. In some embodiments, the invention provides methods of inhibiting MEK activity in a solution by contacting said solution with an amount of a compound of the invention sufficient to inhibit the activity of MEK in said solution. In some embodiments, the invention provides methods of inhibiting MEK activity in a cell by contacting said cell with an amount of a compound of the invention sufficient to inhibit the activity of MEK in said cell.
  • the invention provides methods of inhibiting MEK activity in a tissue by contacting said tissue with an amount of a compound of the invention sufficient to inhibit the activity of MEK in said tissue. In some embodiments, the invention provides methods of inhibiting MEK activity in an organism by contacting said organism with an amount of a compound of the invention sufficient to inhibit the activity of MEK in said organism. In some embodiments, the invention provides methods of inhibiting MEK activity in an animal by contacting said animal with an amount of a compound of the invention sufficient to inhibit the activity of MEK in said animal. In some embodiments, the invention provides methods of inhibiting MEK activity in a mammal by contacting said mammal with an amount of a compound of the invention sufficient to inhibit the activity of MEK in said mammal. In some embodiments, the invention provides methods of inhibiting MEK activity in a human by contacting said human with an amount of a compound of the invention sufficient to inhibit the activity of MEK in said human.
  • This invention also relates to a pharmaceutical composition for inhibiting abnormal cell growth in a mammal which comprises an amount of a compound of the present invention, or a pharmaceutically acceptable salt, ester, solvate or hydrate thereof, in combination with an amount of a chemotherapeutic, wherein the amounts of the compound, salt, ester, solvate or hydrate, and of the chemotherapeutic are together effective in inhibiting abnormal cell growth.
  • chemotherapeutics are presently known in the art and can be used in combination with the compounds of the invention.
  • the chemotherapeutic is selected from the group consisting of mitotic inhibitors, alkylating agents, anti-metabolites, intercalating antibiotics, growth factor inhibitors, cell cycle inhibitors, enzymes, topoisomerase inhibitors, biological response modifiers, anti-hormones, angiogenesis inhibitors, and anti-androgens.
  • This invention further relates to use of a compound of the present invention, or a pharmaceutically acceptable salt, ester, solvate or hydrate thereof, in combination with radiation therapy, in the treatment of inhibiting abnormal cell growth, wherein the amounts of the compound, salt, ester, solvate or hydrate, is in combination with the radiation therapy effective in inhibiting abnormal cell growth.
  • Techniques for administering radiation therapy are known in the art, and these techniques can be used in the combination therapy described herein.
  • the administration of the compound of the invention in this combination therapy can be determined as described herein.
  • the compounds of the present invention can render abnormal cells more sensitive to treatment with radiation for purposes of killing and/or inhibiting the growth of such cells.
  • the invention also relates to use of a compound of the present invention, or a pharmaceutically acceptable salt, ester, solvate or hydrate thereof, or an isotopically-labeled derivative thereof, and an amount of one or more substances selected from anti-angiogenesis agents, signal transduction inhibitors, and antiproliferative agents, in the treatment of inhibiting abnormal cell growth.
  • Anti-angiogenesis agents such as MAP-2 (matrix-metalloprotienase 2) inhibitors, MMP-9 (matrix-metalloprotienase 9) inhibitors, and COX-11 (cyclooxygenase 11) inhibitors, can be used in conjunction with a compound of the present invention and pharmaceutical compositions described herein.
  • useful COX-II inhibitors include CELEBREXTM (alecoxib), valdecoxib, and rofecoxib.
  • Examples of useful matrix metalloproteinase inhibitors are described in WO 96/33172 (published October 24,1996 ), WO 96/27583 (published March 7,1996 ), European Patent Application No.
  • MMP-2 and MMP-9 inhibitors are those that have little or no activity inhibiting MMP-1. More preferred, are those that selectively inhibit MMP-2 and/or AMP-9 relative to the other matrix-metalloproteinases (i.
  • MMP inhibitors useful in the present invention are AG-3340, RO 32-3555, and RS 13-0830.
  • treating and its grammatical equivalents as used herein include achieving a therapeutic benefit and/or a prophylactic benefit.
  • therapeutic benefit is meant eradication or amelioration of the underlying disorder being treated.
  • a therapeutic benefit is achieved with the eradication or amelioration of one or more of the physiological symptoms associated with the underlying disorder such that an improvement is observed in the patient, notwithstanding that the patient may still be afflicted with the underlying disorder.
  • the compositions may be administered to a patient at risk of developing a particular disease, or to a patient reporting one or more of the physiological symptoms of a disease, even though a diagnosis of this disease may not have been made.
  • the compounds of the invention will be co-administer with other agents as described above.
  • the term "Co-administration,” “administered in combination with,” and their grammatical equivalents, as used herein, encompasses administration of two or more agents to an animal so that both agents and/or their metabolites are present in the animal at the same time. Co-administration includes simultaneous administration in separate compositions, administration at different times in separate compositions, or administration in a composition in which both agents are present.
  • the compounds of the invention and the other agent(s) are administered in a single composition.
  • compounds of the invention and the other agent(s) are admixed in the composition.
  • the invention provides methods for synthesizing the compounds described herein.
  • the compounds of this invention can be prepared by the methods described below. The procedures below are intended to illustrate those methods, and the examples given are intended to illustrate the scope of this invention. Neither the methods not the examples should be construed as limiting the invention in any way. The procedures below are explained in more detail in the examples section.
  • the compounds of the present invention may have asymmetric carbon atoms.
  • Diastereomeric mixtures can be separated into their individual diastereomers on the basis of their physical chemical differences by methods known to those skilled in the art, for example, by chromatography or fractional crystallization.
  • Enantiomers can be separated by converting the enantiomer mixture into a diastereomeric mixture by reaction with an appropriate optically active compound (e. g. alcohol), separating the diastereomers and converting (e. g., hydrolyzing) the individual diastereomers to the corresponding pure enantiomers. All such isomers, including diastereomeric mixtures and pure enantiomers are considered as part of the invention.
  • the activity of the compounds of the present invention may be determined by the following procedure, as well as the procedure described in the examples below.
  • N-terminal 6 His-tagged, constitutively active MEK1 (2-393) is expressed in E. coli and protein is purified by conventional methods ( Ahn et al. Science 1994, 265, 966-970 ).
  • the activity of MEK1 is assessed by measuring the incorporation of ⁇ - 33 P-phosphate from ⁇ - 33 P-ATP onto N-terminal His tagged ERK2, which is expressed in E. coli and is purified by conventional methods, in the presence of MEK1.
  • the assay is carried out in 96-well polypropylene plate.
  • the incubation mixture (100, ⁇ L) comprises of 25 mM Hepes, pH 7.4, 10 mM MgCl 2 , 5 mM ⁇ -glycerolphosphate, 100 ⁇ M Na-orthovanadate, 5 mM DTT, 5 nM MEK1, and 1 ⁇ M ERK2.
  • Inhibitors are suspended in DMSO, and all reactions, including controls are performed at a final concentration of 1% DMSO. Reactions are initiated by the addition of 10 ⁇ M ATP (with 0.5 ⁇ Ci ⁇ - 33 P- ATP/well) and incubated at ambient temperature for 45 minutes. Equal volume of 25% TCA is added to stop the reaction and precipitate the proteins.
  • Precipitated proteins are trapped onto glass fiber B filter plates, and excess labeled ATP washed off using a Tomtec MACH III harvestor. Plates are allowed to air-dry prior to adding 30 ⁇ L/well of Packard Microscint 20, and plates are counted using a Packard TopCount.
  • Administration of the compounds of the present invention can be effected by any method that enables delivery of the compounds to the site of action. These methods include oral routes, intraduodenal routes, parenteral injection (including intravenous, subcutaneous, intramuscular, intravascular or infusion), topical, and rectal administration.
  • an effective dosage is in the range of about 0.001 to about 100 mg per kg body weight per day, preferably about 1 to about 35 mg/kg/day, in single or divided doses. For a 70 kg human, this would amount to about 0.05 to 7 g/day, preferably about 0.05 to about 2.5 g/day. In some instances, dosage levels below the lower limit of the aforesaid range may be more than adequate, while in other cases still larger doses may be employed without causing any harmful side effect, e.g. by dividing such larger doses into several small doses for administration throughout the day.
  • the active compound may be applied as a sole therapy or may involve one or more other anti-tumor substances, for example those selected from, mitotic inhibitors, for example vinblastine; alkylating agents, for example cis-platin, carboplatin and cyclophosphamide; anti-metabolites, for example 5-fluorouracil, cytosine arabinside and hydroxyurea, or, for example, one of the preferred anti-metabolites disclosed in European Patent Application No.
  • mitotic inhibitors for example vinblastine
  • alkylating agents for example cis-platin, carboplatin and cyclophosphamide
  • anti-metabolites for example 5-fluorouracil, cytosine arabinside and hydroxyurea, or, for example, one of the preferred anti-metabolites disclosed in European Patent Application No.
  • Such conjoint treatment may be achieved by way of the simultaneous, sequential or separate dosing of the individual components of treatment.
  • the pharmaceutical composition may, for example, be in a form suitable for oral administration as a tablet, capsule, pill, powder, sustained release formulations, solution, suspension, for parenteral injection as a sterile solution, suspension or emulsion, for topical administration as an ointment or cream or for rectal administration as a suppository.
  • the pharmaceutical composition may be in unit dosage forms suitable for single administration of precise dosages.
  • the pharmaceutical composition will include a conventional pharmaceutical carrier or excipient and a compound according to the invention as an active ingredient. In addition, it may include other medicinal or pharmaceutical agents, carriers, adjuvants, etc.
  • Exemplary parenteral administration forms include solutions or suspensions of active compounds in sterile aqueous solutions, for example, aqueous propylene glycol or dextrose solutions. Such dosage forms can be suitably buffered, if desired.
  • Suitable pharmaceutical carriers include inert diluents or fillers, water and various organic solvents.
  • the pharmaceutical compositions may, if desired, contain additional ingredients such as flavorings, binders, excipients and the like.
  • excipients such as citric acid
  • disintegrants such as starch, alginic acid and certain complex silicates
  • binding agents such as sucrose, gelatin and acacia.
  • lubricating agents such as magnesium stearate, sodium lauryl sulfate and talc are often useful for tableting purposes.
  • Solid compositions of a similar type may also be employed in soft and hard filled gelatin capsules.
  • Preferred materials include lactose or milk sugar and high molecular weight polyethylene glycols.
  • active compound may be combined with various sweetening or flavoring agents, coloring matters or dyes and, if desired, emulsifying agents or suspending agents, together with diluents such as water, ethanol, propylene glycol, glycerin, or combinations thereof.
  • kits for the treatment of disorders such as the ones described herein.
  • kits comprise a compound or compounds described herein in a container and, optionally, instructions teaching the use of the kit according to the various methods and approaches described herein.
  • kits may also include information, such as scientific literature references, package insert materials, clinical trial results, and/or summaries of these and the like, which indicate or establish the activities and/or advantages of the composition, and/or which describe dosing, administration, side effects, drug interactions, or other information useful to the health care provider..
  • Such information may be based on the results of various studies, for example, studies using experimental animals involving in vivo models and studies based on human clinical trials.
  • Kits described herein can be provided, marketed and/or promoted to health providers, including physicians, nurses, pharmacists, formulary officials, and the like. Kits may also, in some embodiments, be marketed directly to the consumer.
  • Fuming nitric acid (1.7 ml) is added dropwise to concentrated sulfuric acid (25 ml) while maintaining the temperature at 5 - 10 °C.
  • 2,3,4-Trifluorobenzoic acid (5 g, 28 mmoles) is added in small portion to this solution while keeping the reaction temperature at 5 °C.
  • the reaction mixture is stirred at room temperature for an additional 2 hours and poured into ice.
  • the mixture is extracted with ether (3 x 75 ml). The organic layers are combined, washed with brine, dried (MgSO 4 ). The solvent is removed, and the crude product is recrystallized from hot chloroform to obtain the title compound.
  • Step B 3,4-Difluoro-2-(2-fluoro-4-iodophenylamino)-5-nitrobenzoic acid:
  • Step C Methyl 3,4-difluoro-2-(2-fluoro-4-iodophenylamino)-5-nitrobenzoate:
  • Step D Methyl 4-(allylamino)-3-fluoro-2-(2-fluoro-4-iodophenylamino)-5-nitrobenzoate:
  • Step E Methyl 4-(allylamino)-5-amino-3-fluoro-2-(2-fluoro-4-iodophenylamino)benzoate:
  • Step F Methyl 1-allyl-7-fluoro-6-(2-fluoro-4-iodophenylamino)-1H-benzo[d]imidazole-5-carboxylate:
  • Step G Methyl 3-allyl-4-fluoro-5-(3-fluoro-4-iodophenylamino)-6-methoxycarbony-1-methyl-3H-benzoimidazol-1-ium iodide:
  • Step H Methyl 4-fluoro-5-(3-fluoro-4-iodophenylamino)-1-methyl-1H-benzo[d]imidazole-6-carboxylate:
  • Step I 4-Fluoro-5-(2-fluoro-4-iodophenylamino)-1-methyl-1H-benzo[d]imidazole-6-carboxylic acid:
  • Methyl 4-fluoro-5-(2-fluoro-4-iodophenylamino)-1-methyl-1H-benzo[d]imidazole-6-carboxylate (2.15 g, 4.8 mmoles) is dissolved in a mixture of THF (30 ml), MeOH (10 ml) and H 2 O (5 ml) and LiOH (1.16 g, 4.8 mmoles) is added. The mixture is stirred at room temperature for 2 hours. Solvents are removed under reduced pressure and 1N HCl is added. The aqueous layer is extracted with chloroform. The organic layers are combined, washed with brine, dried (Na 2 SO 4 ) and concentrated under reduced pressure to obtain the title compound.
  • Step J 1-(2-fluoro-4-iodophenylamino)-8-fluoro-5-methyl-benzimidazo[6,5-d]imidazole:
  • Step K 3-cyclopropanesulfonamide 1-(2-fluoro-4-iodophenylamino)-8-fluoro-5-methyl-benzimidazo[6,5-d]imidazole:
  • Step J The previous compound (Step J) (50 mg, 0.117 mmoles) is dissolved in THF (2 ml) and the mixture is cooled at -78 °C. LiHMDS (0.117 ml, 0.117 mmoles) is added dropwise followed by the addition of HMPA (0.5 ml). The mixture is warmed at room temperature and cyclopropyl sulfonyl chloride (0.013 ml, 0.14 mmoles) is added. After stirring at room temperature overnight, a saturated solution of ammonium chloride (5 ml) is added. The mixture is extracted with EtOAc (3 x 5 ml). The organic layers are combined, washed with brine, dried (Na 2 SO 4 ) and concentrated under reduced pressure. The crude product is purified by flash chromatography using a gradient of EtOAc and hexane yielding to the title compound.
  • Step L N-(4-fluoro-5-(3-fluoro-4-iodophenylamino)-1-methyl-1H-benzo[d]imidazol-6-yl)cyclopropanesulfonamide:
  • Step K The previous compound (Step K) (26 mg, 0.049 mmoles), potassium trimethylsilanolate (21 mg, 0.149 mmoles) dissolved in THF (2 ml) are stirred at room temperature overnight. Brine (3ml) is added and the mixture is extracted with EtOAc (3 x 5 ml). The organic layers are combined, dried (Na 2 SO 4 ) and concentrated under reduced pressure. The crude product is purified by flash chromatography using a gradient of EtOAc and hexane yielding to the title compound.
  • Step B (3-(1-bromocyclopropyl)ethoxy)(tert-butyl)dimethylsilane:
  • Step C 1-(2-(tert-butyldimethylsilyloxy)ethyl)cyclopropane-1-sulfonyl chloride:
  • step B To a solution of the cyclopropyl bromide prepared in step B (1.1227 g, 4.04 mmoles) in 15 ml anhydrous diethyl ether was added a 1.7 M solution of t-BuLi in pentane (4.8 ml, 8.16 mmoles) at - 78 °C. The solution was stirred for 30 min at this temperature, and was then transferred via a transfer cannula into a solution of freshly distilled sulfuryl chloride (0.65 ml, 1.029 g, 8.1 mmoles) in 8 ml diethyl ether at - 78 °C. The yellow suspension was warmed to room temperature.
  • Step D 1-(2-(tert-butyldimethylsilyloxy)ethyl)-1-(2-fluoro-4-iodophenylamino)-8-fluoro-5-methyl-benzimidazo[6,5-d]imidazolecyclopropane-3-sulfonamide:
  • step K 1-(3-fluoro-4-iodophenylamino)-8-fluoro-5-methyl-benzimidazo[6,5-d]imidazole (81 mg, 0.19 mmoles) was reacted with the cyclopropylsulfonyl chloride (162 mg, 0.38 mmoles) prepared in step C to obtain the title product (46.6 mg, 36%).
  • m / z 687 [M-1] - .
  • Step E 1-(2-(Tert-butyldimethylsilyloxy)ethyl)-N-(4-fluoro-5-(2-fluoro-4-iodophenylamino)-1-methyl-1H-benzo[d]imidazol-6-yl)cyclopropane-1-sulfonamide:
  • step L the previous compound (46.6 mg, 0.067 mmoles) was reacted with potassium trimethylsilanolate (26 mg, 0.230 mmoles) in THF (1.5 ml) to obtain the title compound (39 mg, 87%).
  • Step F N-(4-fluoro-5-(2-fluoro-4-iodophenylamino)-1-methyl-1H-benzo[d]imidazol-6-yl)-1-(2-hydroxyethyl)cyclopropane-1-sulfonamide:
  • Step A 5-Bromo-3,4-difluoro-3-(3-fluorophenylamino)benzoic acid:
  • Step B Methyl 5-bromo-3,4-difluoro-2-(2-fluorophenylamino)benzoate:
  • Step C Methyl 3,4-difluoro-3-(3-fluorophenylamino)-5-((trimethylsilyl)ethynyl)benzoate:
  • Step D Methyl 5-acetyl-3,4-difluoro-2-(2-fluorophenylamino)benzoate:
  • Step E Methyl 7-fluoro-6-(3-fluorophenylamino)-3-methylbenzo[d]isoxazole-5-carboxylate:
  • the reaction was quenched with saturated aqueous NH 4 Cl solution (200 ml) and extracted with EtOAc (2 x 200 ml). The organic solution was washed with brine (100 ml), dried over MgSO 4 and concentrated. To the resulting oil was added a mixture of 5% HCl solution (100 ml) and MeOH (100 ml). The resulting suspension was heated at reflux for 1.5 h. The reaction was cooled to room temperature, concentrated to half volume, and diluted with EtOAc (200 ml). The organic solution was washed with brine (100 ml), dried over Na 2 SO 4 and concentrated.
  • Step F Methyl 7-fluoro-6-(2-fluoro-4-iodophenylamino)-3-methylbenzo[d]isoxazole-5-carboxylate:
  • Step G 7-Fluoro-6-(2-fluoro-4-iodophenylamino)-3-methylbenzo[d]isoxazole-5-carboxylic acid:
  • Step H 8-fluoro-1-(2-fluoro-4-iodophenyl)-5-methylbenzimidazolo[6,5-d][d]isoxazole:
  • Step I 3-(Cyclopropanesulfonyl)-8-fluoro-1-(2-fluoro-4-iodophenyl)-5-methylbenzimidazolo[6,5-d][d]isoxazole:
  • Step J N-(7-fluoro-6-(3-fluoro-4-iodophenylamino)-3-methylbenzo[d]isoxazol-5-yl)cyclopropanesulfonamide:
  • Step A 3-(3-( tert -butyldimethylsilyloxy)ethyl)cyclopropanesulfonyl)-8-fluoro-1-(3-fluoro-4-iodophenyl)-5-methylbenzimidazolo[6,5-d][d]isoxazole:
  • step H 8-fluoro-1-(2-fluoro-4-iodophenyl)-5-methylbenzimidazolo[6,5-d][d]isoxazole (example 3, step H) (150 mg, 0.35 mmol) in THF (15 ml) at -78 °C was added LiHMDS (0.39 ml, 1 M in THF, 0.39 mmol). The reaction mixture was stirred at -78 °C for 30 min and 1-(2-(tert-butyldimethylsilyloxy)ethyl)cyclopropane-1-sulfonyl chloride (144 mg, 0.48 mmol) was added to the mixture.
  • the reaction was slowly warmed to room temperature and stirred at the temperature for 16 h.
  • the reaction was quenched with saturated aqueous NH 4 Cl solution (20 ml) and extracted with EtOAc (2 x 30 ml). The combined organic solution was washed with brine (30 ml), dried over MgSO 4 and concentrated.
  • Step B 1-(3-( tert -butyldimethylsilyloxy)ethyl)-N-(7-fluoro-6-(3-fluoro-4-iodophenylamino)-3-methylbenzo[d]isoxazol-5-yl)cyclopropane-1-sulfonamide:
  • Step C 1-(3-hydroxyethyl)-N-(7-fluoro-6-(2-fluoro-4-iodophenylamino)-3-methylbenzo[d]isoxazol-5-yl)cyclopropane-1-sulfonamide:
  • Step A 3-(allylcyclopropanesulfonyl)-8-fluoro-1-(2-fluoro-4-iodophenyl)-5-methylbenzimidazolo[6,5- d][d]isoxazole:
  • step H To a solution of 8-fluoro-1-(2-fluoro-4-iodophenyl)-5-methylbenzimidazolo[6,5-d][d]isoxazole (example 3, step H) (300 mg, 0.70 mmol) in THF (30 ml) at -78 °C was added LiHMDS (0.77 ml, 1 M in THF, 0.77 mmol). The reaction mixture was stirred at -78 °C for 30 min and 1-allylcyclopropane-1-sulfonyl chloride (253 mg, 1.40 mmol) was added to the mixture. The reaction was slowly warmed to room temperature and stirred at the temperature for 3 days.
  • Step B 1-allyl-N-(7-fluoro-6-(2-fluoro-4-iodophenylamino)-3-methylbenzo[d]isoxazol-5-yl)cyclopropane-1-sulfonamide:
  • Step C 1-(2,3-dihydroxypropyl)-N-(7-fluoro-6-(2-fluoro-4-iodophenylamino)-3-methylbenzo[d]isoxazol-5-yl)cyclopropane-1-sulfonamide:
  • Step A 8-fluoro-1-(4-bromo-2-chlorophenyl)-5-methylbenzimidazolo[6,5-d][d]isoxazole:
  • Step B 3-(Cyclopropanesulfonyl)-8-fluoro-1-(4-bromo-2-chlorophenyl)-5-methylbenzimidazolo[6,5-d][d]isoxazole:
  • Step C N-(7-fluoro-6-(4-bromo-2-chlorophenylamino)-3-methylbenzo[d]isoxazol-5-yl)cyclopropanesulfonamide:
  • Step A Methyl 7-fluoro-6-(2-fluorophenylamino)-3-methylbenzo[c]isoxazole-5-carboxylate:
  • step D To a solution of methyl 5-acetyl-3,4-difluoro-2-(2-fluorophenylamino)benzoate (example 3, step D) (1.50 g, 4.64 mmol) in a mixture of acetone (36 ml) and H 2 O (12 ml) was added NaN 3 (452 mg, 6.96 mmol). The reaction was heated at 65 °C for 16 h and cooled to room temperature. Acetone was concentrated and the resulting mixture was extracted with EtOAc (2 x 50 ml). The combined organic solution was washed with brine (50 ml), dried over MgSO 4 and concentrated to give a yellow solid.
  • Step B methyl 7-fluoro-6-(2-fluoro-4-iodophenylamino)-3-methylbenzo[c]isoxazole-5-carboxylate:
  • Step C 7-fluoro-6-(2-fluoro-4-iodophenylamino)-3-methylbenzo[c]isoxazole-5-carboxylic acid:
  • Step D 8-fluoro-1-(2-fluoro-4-iodophenyl)-5-methylbenzimidazolo[6,5-d][c]isoxazole:
  • Step E 3-(cyclopropanesulfonyl)-8-fluoro-1-(2-fluoro-4-iodophenyl)-5-methylbenzimidazolo[6,5-d][c]isoxazole:
  • Step F N-(7-fluoro-6-(3-fluoro-4-iodophenylamino)-3-methylbenzo[c]isoxazol-5-yl)cyclopropanesulfonamide:
  • This compound is synthesized from 8-fluoro-1-(2-fluoro-4-iodophenyl)-5-methylbenzimidazolo[6,5-d][c]isoxazole according to example 4 (step A, B and C).
  • Step B 4,6-Dichloronicotinic acid:
  • Step C 4-(4-Bromo-2-chlorophenylamino)-6-chloronicotinic acid:
  • Step D 4-(4-Bromo-3-chlorophenylamino)-5,6-dichloronicotinic acid:
  • Step E Methyl 4-(4-bromo-2-chlorophenylamino)-5,6-dichloronicotinate:
  • Step F Methyl 6-azido-4-(4-bromo-3-chlorophenylamino)-5-chloronicotinate:
  • Methyl 4-(4-bromo-2-chlorophenylamino)-5,6-dichloronicotinate (1.690 g, 4.14 mmol) was suspended in 30 ml DMF and the suspension was heated until everything went into solution. It was cooled down to room temperature and to that solution was added solid sodium azide (0.533 g, 8.2 mmol). The yellow solution was stirred for 17 h at room temperature and was diluted with water. The precipitate was filtered, washed with water and hexanes and dried in oil-pump vacuo at 50 °C for 2 h to obtain the title compound as a light-brown solid (1.5422 g, 90%).
  • Step G Methyl 6-amino-4-(4-bromo-2-chlorophenylamino)-5-chloronicotinate:
  • Step H Methyl 7-(4-bromo-2-chlorophenylamino)-8-chloroimidazo[1,2-a]pyridine-6-carboxylate:
  • Step I 7-(4-Bromo-2-chlorophenylamino)-8-chloroimidazo[1,2-a]pyridine-6-carboxylic acid:
  • Step J 3-(4-Bromo-3-chlorophenyl)-4-chloro-1H-diimidazo[1,3-a:4',5'-d]pyridin-2(3H)-one:
  • Step K 3-(4-bromo-3-chlorophenyl)-4-chloro-1-(cyclopropylsulfonyl)-1H-diimidazo[1,2-a:4',5'-d]pyridin-2(3H)-one:
  • Step L N-(7-(4-bromo-3-chlorophenylamino)-8-chloroimidazo[1,2-a]pyridin-6-yl)cyclopropanesulfonamide:
  • This compound was synthesized from 3-(4-bromo-2-chlorophenyl)-4-chloro-1-(cyclopropylsulfonyl)-1H-diimidazo[1,2-a:4',5'-d]pyridin-2(3H)-one (see example 9) according to example 4 (step A, B and C).
  • This compound is synthesized from methyl 6-amino-5-chloro-4-(2-fluoro-4-iodophenylamino]nicotinate: (see below) according to example 9 (step H through L).
  • Step A 6-Dichloro-4-(2-fluoro-4-iodophenylamino)nicotinic acid:
  • Step B 5,6-Dichloro-4-(2-fluoro-4-iodophenylamino)nicotinic acid:
  • Step C Methyl 5,6-dichloro-4-(2-fluoro-4-iodophenylamino)nicotinate:
  • This compound is synthesized from 5,6-dichloro-4-(2-fluoro-4-iodophenylamino)nicotinic acid (3.00 g, 7.64 mmol) according to example 9, step E.
  • Step D Methyl 6-azido-5-chloro-4-(2-fluoro-4-iodophenylamino)nicotinate:
  • This compound is synthesized from methyl 5,6-dichloro-4-(2-fluoro-4-iodophenylamino)nicotinate (2.9 g, 6.5 mmol) according to example 9, step F.
  • Step E Methyl 6-amino-5-chloro-4-(2-fluoro-4-iodophenylamino)nicotinate:
  • This compound is synthesized from methyl 6-azido-5-chloro-4-(2-fluoro-4-iodophenylamino)nicotinate (2.65 g, 5.9 mmol) according to example 9, step G.
  • This compound is synthesized according to example 9 (step H through L) starting from methyl 6-amino-5-fluoro-4-(2-fluoro-4-iodophenylamino)nicotinate (see below).
  • step E To a solution of methyl 6-amino-5-chloro-4-(2-fluoro-4-iodophenylamino)nicotinate (2.0 g, 4.74 mmol) (Example 11, step E) in a mixture of MeOH/water (1/1) (20 ml) is added selectfluor (1.6 g, 4.74 mmol). The mixture is stirred at room temperature overnight, diluted with EtOAc and washed with 0.5 N HCl and brine. The organic extracts are dried (Na 2 SO 4 ) and concentrated under reduced pressure. The crude material was purified by HPLC (reverse phase) to obtain the title compound.
  • This compound is synthesized according to example 4 (step A, B and C) starting from 3-(2-fluoro-4-iodophenyl)-4-methyl-1H-diimidazo[1,2-a:4',5'-d]pyridin-2(3H)-one.
  • step A, B and C This compound was synthesized according to example 5 (step A, B and C) starting from 3-(2-fluoro-4-iodophenyl)-4-methyl-1H-diimidazo[1,2-a:4',5'-d]pyridin-2(3H)-one.
  • m / z 559 [M-1] - .
  • Example 19 N-(8-chloro-7-(2-fluoro-4-iodophenylamino)-3-methyl-[1,2,4]diazole[4,3-a]pyridin-6-yl)cyclopropanesulfonamide:
  • Step A 5-chloro-4-(2-fluoro-4-iodophenylamino)hydrazinylnicotinic acid:
  • Step B 8-Chloro-7-(2-fluoro-4-iodophenylamino)-3-methyl-[1,2,4]triazolo[4,3-a]pyridine-6-carboxylic acid:
  • Acetic anhydride (0.95 mmol) is added to a solution of 5-chloro-4-(2-fluoro-4-iodophenylamino)-6-hydrazinylnicotinic acid (0.4 g, 0.95 mmol) and triethylamine (1.9 mmol) in DCM (10 ml) at 0 °C.
  • the reaction mixture is warmed to room temperature, stirred for an additional 10 min and POCl 3 (0.95 mmol) is added. After stirring overnight at room temperature the mixture is refluxed for 3 days. It was diluted with EtOAc and NaHCO 3 and stirred for 20 min. The organic layer is washed with H 2 O, brine, dried with Na 2 SO 4 and concentrated under reduced pressure.
  • the crude product is purified by column chromatography.
  • Step C 9-chloro-8-(2-fluoro-4-iodophenyl)-3-methyl-6H-imidazo][4,5-d][1,2,4]triazolo[4,3-a]pyridin-7(8H)-one:
  • step H from 9-chloro-8-(2-fluoro-4-iodophenyl)-3-methyl-6H-imidazo[4,5-d][1,2,4]triazolo[4,3-a]pyridin-7(8H)-one (0.34, 0.76 mmol) to afford the title compound.
  • Step D 9-chloro-6-(cyclopropylsulfonyl)-8-(2-fluoro-4-iodophenyl)-3-methyl-6H-imidazo[4,5-d][1,2,4]triazolo[4,3-a]pyridin-7(8H)-one:
  • step I from 9-chloro-8-(2-fluoro-4-iodophenyl)-3-methyl-6H-imidazo[4,5-d][1,2,4]triazolo[4,3-a]pyridin-7(8H)-one (100 mg, 0.22 mmol) to afford the title compound.
  • Step E N-(8-chloro-7-(2-fluoro-4-iodophenylamino)-3-methyl-[1,2,4]triazolo[4,3-a]pyridin-6-yl)cyclopropanesulfonamide:
  • step J from 9-chloro-6-(cyclopropylsulfonyl)-8-(2-fluoro-4-iodophenyl)-3-methyl-6H-imidazo[4,5-d][1,2,4]triazolo[4,3-a]pyridin-7(8H)-one (98 mg, 018 mmol) to afford the title compound.
  • Example 20 N-(8-chloro-7-(2-fluoro-4-iodophenylamino)-3-methyl-[1,2,4]triazolo[4,3-a]pyridin-6-yl)-1-(2-hydroxyethyl)cyclopropane-1-sulfonamide:
  • step A, B and C from 9-chloro-8-(2-fluoro-4-iodophenyl)-3-methyl-6H-imidazo[4,5-d][1,2,4]triazolo[4,3-a]pyridin-7(8H)-one to afford the title compound.
  • Example 21 N-(7-chloro-6-(2-fluoro-4-iodophenylamino)-3-methyl-3a,7a-dihydro-3H-imidazo[4,5-b]pyridin-5-yl)cyclopropanesulfonamide:
  • Step A 7-Chloro-6-(2-fluoro-4-iodophenylamino)-3-methyl-3H-imidazo[4,5-b]pyridine-5-carboxylic acid:
  • Step B 8-Chloro-1-(3-fluoro-4-iodophenyl)-5-methyl-3,5-dihydrodiimidazo[4,5-b:4',5'-e]pyridin-3(1H)-one:
  • Step C 8-Chloro-3-(cyclopropylsulfonyl)-1-(2-fluoro-4-iodophenyl)-5-methyl-3,4a,5,7a-tetrahydrodiimidazo[4,5-b:4',5'-e]pyridin-2(1H)-one:
  • Step D N-(7-Chloro-6-(2-fluoro-4-iodophenylamino)-3-methyl-3a,7a-dihydro-3H-imidazo[4,5-b]pyridin-5-yl)cyclopropanesulfonamide:
  • Example 22 N-(7-chloro-6-(2-fluoro-4-iodophenylamino)-3-methyl-3a,7a-dihydro-3H-imidazo[4,5-b]pyridin-5-yl)-1-(2-hydroxyethyl)cyclopropane-1-sulfonamide:
  • Step A 3-(1-(2-(Tert-butyldimethylsilyloxy)ethyl)cyclopropylsulfonyl)-8-chloro-1-(2-fluoro-4-iodophenyl)-1-5-methyl-3,4a,5,7a-tetrahydrodiimidazo[4,5-b:4',5'-e]pyridin-2(1H)-one:
  • reaction mixture is stirred at -78 °C for 30 min and 1-(2-(tert-butyldimethylsilyloxy)ethyl)cyclopropane-1-sulfonyl chloride (144 mg, 0.48 mmol) is added to the mixture.
  • the reaction is slowly warmed to room temperature and stirred at the temperature for 16 h.
  • the reaction is quenched with saturated aqueous NH 4 Cl solution (20 ml) and extracted with EtOAc (2 x 30 ml). The combined organic solution is washed with brine (30 ml), dried over MgSO 4 and concentrated.
  • Silica gel chromatography yields the desired product.
  • Step B 1-(2-(Tert-butyldimethylsilyloxy)ethyl)-N-(7-chloro-6-(2-fluoro-4-iodophenylamino)-3-methyl-3a,7a-dihydro-3H-imidazo[4,5-b]pyridin-5-yl)cyclopropane-1-sulfonamide:
  • Step C N-(7-chloro-6-(2-fluoro-4-iodophenylamino)-3-methyl-3a,7a-dihydro-3H-imidazo[4,5-b]pyridin-5-yl)-1-(2-hydroxyethyl)cyclopropane-1-sulfonamide:
  • Example 23 N-(7-(4-bromo-2-fluorophenylamino)imidazo[1,2-b]pyridazin-6-yl)cyclopropanesulfonamide:
  • Step A Preparation of 4,6-dichloro-pyridazine-3-carboxylic acid:
  • Step B 4-(4-Bromo-2-fluoro-phenylamino)-6-chloro-pyridazine-3-carboxylic acid:
  • the reaction is quenched by addition of 5 ml of water, diluted with ethyl acetate, acidified with aqueous 1.0 M HCl to pH 1-2.
  • the layers are separated and aqueous layer is extracted with ethyl acetate (30 ml x 3).
  • the combined organic layers are washed with brine (30 ml x 3), dried over Na 2 SO 4 , and concentrated under reduced pressure.
  • the residue is purified by flash column chromatography to give the product.
  • Step C 4-(4-Bromo-2-fluoro-phenylamino)-6-chloro-pyridazine-3-carboxylic acid tert-butyl ester:
  • Step D 6-Azido-4-(4-Bromo-2-fluoro-phenylamino)-pyridazine-3-carboxylic acid tert-butyl ester:
  • Step E 6-Amino-4-(4-Bromo-2-fluoro-phenylamino)-6-chloro-pyridazine-3-carboxylic acid tert-butyl ester:
  • Step F 7-(4-Bromo-2-fluoro-phenylamino)-imidazo[1,2-b]pyridazine-6-carboxylic acid:
  • Step G 1-(4-Bromo-2-fluoro-phenyl)-1H,3H-1,3,4,4a,7-pentaaza-s-indacen-2-one:
  • step B from 7-(4-bromo-2-fluoro-phenylamino)-imidazo[1,2-b]pyridazine-6-carboxylic acid to afford the title compound.
  • Step H 1-(4-Bromo-2-fluoro-phenyl)-3-(butane-2-sulfonyl)-1H,3H-1,3,4,4a,7-pentaaza-s-indacen-2-one:
  • step C from 1-(4-bromo-2-fluoro-phenyl)-1H,3H-1,3,4,4a,7-pentaaza-s-indacen-2-one to afford the title compound.
  • Step I Cyclopropanesulfonic acid [7-(4-bromo-2-fluoro-phenylamino)-imidazo[1,2-b]pyridazin-6-yl]-amide:
  • step D from 1-(4-bromo-2-fluoro-phenyl)-3-(butane-2-sulfonyl)-1H,3H-1,3,4,4a,7-pentaaza-s-indacen-2-one to afford the title compound.
  • Example 24 N-(7-(4-bromo-2-fluorophenylamino)imidazo[1,2-b]pyridazin-6-yl)-1-(2-hydroxyethyl)cyclopropane-1-sulfonamide:
  • step A, B and C from 1-(4-bromo-2-fluoro-phenyl)-1H,3H-1,3,4,4a,7-pentaaza-s-indacen-2-one to afford the title compound.
  • Example 25 N-(7-fluoro-6-(2-fluoro-4-iodophenylamino)pyrazolo[1,5-a]pyrimidin-5-yl)cyclopropanesulfonamide:
  • This compound is synthesized according to example 21 (step A through D) starting from 6-chloro-7-fluoropyrazolo[1,5-a]pyrimidine-5-carboxylic acid ( WO 2005/051906 ).
  • Example 26 N-(7-fluoro-6-(2-fluoro-4-iodophenylamino)pyrazolo[1,5-a]pyrimidin-5-yl)-1-(2-hydroxyethyl)cyclopropane-1-sulfonamide:
  • This compound is synthesized according to example 22 (step A through C) starting from 3-(cyclopropylsulfonyl)-9-fluoro-1-(2-fluoro-4-iodophenyl)-1H-pyrazolo[1,5-a]purin-2(3H)-one (example 25).
  • Example 27 N-(6-(4-bromo-2-fluorophenylamino)-7-fluoro-3-methylisoxazolo[4,5-b]pyridin-5-yl)cyclopropanesulfonamide:
  • This compound is synthesized according to example 4 (step B through D) starting from 6-(4-bromo-2-fluorophenylamino)-7-fluoro-3-methylisoxazolo[4,5-b]pyridine-5-carboxylic acid ( WO 2005/051906 ).
  • Example 28 N-(6-(4-bromo-2-fluorophenylamino)-7-fluoro-3-methylisoxazolo[4,5-b]pyridin-5-yl)-1-(2-hydroxyethyl)cyclopropane-1-sulfonamide:
  • Example 29 N-(6-(2-fluoro-4-iodophenylamino)-7-fluoro-2,3-dimethyl-2H-indazol-5-yl)cyclopropanesulfonamide:
  • Step A 7-fluoro-6-(2-fluoro-phenylamino)-3-methyl-2H-indazole-5-carboxylic acid methyl ester:
  • Step B 7-fluoro-6-(2-fluoro-phenylamino)-2,3-dimethyl-2H-indazole-5-carboxylic acid methyl ester:
  • Step C 7-fluoro-6-(2-fluoro-4-iodophenylamino)-2,3-dimethyl-2H-indazole-5-carboxylic acid methyl ester:
  • Step D 7-fluoro-6-(2-fluoro-4-iodophenylamino)-2,3-dimethyl-2H-indazole-5-carboxylic acid:
  • Step E 8-fluoro-7-(2-fluoro-4-iodophenyl)-2,3-dimethyl-2,7-dihydro-5H-imidazo[4,5-f]indazol-6-one:
  • Step F 5-cyclopropanesulfonyl-8-fluoro-7-(2-fluoro-4-iodophenyl)-2,3-dimethyl-2,7-dihydro-5H-imidazo[4,5-f]indazol-6-one:
  • Step G N-(6-(2-fluoro-4-iodophenylamino)-7-fluoro-2,3-dimethyl-3H-indazol-5-yl)cyclopropanesulfonamide:
  • Example 30 N-(6-(2-fluoro-4-iodophenylamino)-7-fluoro-1,3-dimethyl-1H-indazol-5-yl)cyclopropanesulfonamide:
  • step C through G This compound was synthesized according to example 29 (step C through G) starting from 7-fluoro-6-(2-fluoro-phenylamino)-1,3-dimethyl-1H-indazole-5-carboxylic acid methyl ester (step B, example 29).
  • m / z 517 [M-1] - .
  • Example 31 N-(6-(2-fluoro-4-iodophenylamino)-7-fluoro-2,3-dimethyl-2H-indazol-5-yl)-1-(2-hydroxyethyl)cyclopropane-1-sulfonamide:
  • This compound is synthesized according to example 4 (step A through C) starting from 8-fluoro-7-(2-fluoro-4-iodophenyl)-2,3-dimethyl-2,7-dihydro-5H-imidazo[4,5-f]indazol-6-one (example 29).
  • Step A Methyl 7-fluoro-6-(2-fluorophenylamino)-3-methylisoxazolo[4,3-b]pyridine-5-carboxylate:
  • Step B Methyl 7-fluoro-6-(2-fluoro-4-iodophenylamino)-3-methylisoxazolo[4,3-b]pyridine-5-carboxylate:
  • Step C 7-Fluoro-6-(2-fluoro-4-iodophenylamino)-3-methylisoxazolo[4,3-b]pyridine-5-carboxylic acid:
  • Step D 8-Fluoro-7-(2-fluoro-4-iodophenyl)-3-methyl-5H-imidazo[4,5-b]isoxazolo[3,4-e]pyridin-6(7H)-one:
  • Step E 5-(Cyclopropylsulfonyl)-8-fluoro-7-(2-fluoro-4-iodophenyl)-3-methyl-5H-imidazo[4,5-b]isoxazolo[3,4 - e]pyridin-6 ( 7H)-one :
  • Step F N-(7-fluoro-6-(2-fluoro-4-iodophenylamino)-3-methylisoxazolo[4,3-b]pyridin-5-yl)cyclopropanesulfonamide:
  • Example 34 (not claimed ) : N-(7-(2-fluoro-4-iodophenylamino)-8-fluoro-4-methylquinazolin-6-yl)cyclopropanesulfonamide:
  • the orange solution was stirred at -78 °C for 1 h, then transferred, via cannula, to freshly grinded dry CO 2 .
  • the reaction was warmed up to room temperature and stirred overnight.
  • the reaction mixture was cooled at 0 °C, quenched with 10% HCl (300 ml), extracted with ether (100 ml x 3), dry over MgSO 4 and concentrated under reduced pressure to obtain (10.6 g, 88%) of the title compound.
  • Step B 5-Bromo-2-(2-fluorophenylamino)-3,4-difluorobenzoic acid:
  • Step D Methyl 2-(2-fluorophenylamino)-3,4-difluoro-5-((trimethylsilyl)ethynyl)benzoate:
  • Step E Methyl 5-acetyl-2-(2-fluorophenylamino)-3,4-difluorobenzoate :
  • Step F Methyl 5-acetyl-2-(2-fluoro-4-iodophenylamino)-3,4-difluorobenzoate :
  • Step G Methyl 7-(2-fluoro-4-iodophenylamino)-8-fluoro-4-methylquinazoline-6-carboxylate:
  • Step H 7-(2-Fluoro-4-iodophenylamino)-8-fluoro-4-methylquinazoline-6-carboxylic acid:
  • Step I 3-(2-Fluoro-4-iodophenyl)-4-fluoro-8-methyl-1H-imidazo[4,5-g]quinazolin-2(3H)-one:
  • Step J 3-(2-Fluoro-4-iodophenyl)-1-(cyclopropylsulfonyl)-4-fluoro-8-methyl-1H-imidazo[4,5-g]quinazolin-2(3H)-one:
  • Step K N-(7-(2-Fluoro-4-iodophenylamino)-8-fluoro-4-methylquinazolin-6-yl)cyclopropanesulfonamide:
  • Example 35 N-(7-(2-fluoro-4-iodophenylamino)-8-fluoro-4-methylquinazolin-6-yl)-1-(2-hydroxyethyl)cyclopropane-1-sulfonamide:
  • Step A 3-(2-Fluoro-4-iodophenyl)-1-(1-(2-(tert-butyldimethylsilyloxy)ethyl)cyclopropylsulfonyl)-4-fluoro-8-methyl-1H-imidazo[4,5-g]quinazolin-2(3H)-one:
  • Step B N-(7-(2-Fluoro-4-iodophenylamino)-8-fluoro-4-methylquinazolin-6-yl)-1-(2-(tert-butyldimethylsilyloxy)ethyl)cyclopropane-1-sulfonamide:
  • Step C N-(7-(2-Fluoro-4-iodophenylamino)-8-fluoro-4-methylquinazolin-6-yl)-1-(2-hydroxyethyl)cyclopropane-1-sulfonamide:
  • Example 36 (not claimed ) : N-(7-(2,4-dichlorophenylamino)-8-fluoro-4-methylcinnolin-6-yl)cyclopropanesulfonamide:
  • This compound is synthesized according to example 34 starting from 7-(4-bromo-2-chlorophenylamino)-8-fluoro-4-methylcinnoline-6-carboxylic acid ( WO 2005/051302 ).
  • Example 37 N-(7-(2,4-dichlorophenylamino)-8-fluoro-4-methylcinnolin-6-yl)-1-(2-hydroxyethyl)cyclopropane-1-sulfonamide:
  • Example 38 N-(7-(4-bromo-2-fluorophenylamino)-8-fluoro-4-methylquinolin-6-yl)cyclopropanesulfonamide:
  • This compound is synthesized according to example 34 starting from 7-(4-bromo-2-fluorophenylamino)-8-fluoro-4-methylquinoline-6-carboxylic acid ( WO 2005/051302 ).
  • Example 39 N-(7-(4-bromo-2-fluorophenylamino)-8-fluoro-4-methylquinolin-6-yl)-1-(2-hydroxyethyl)cyclopropane-1-sulfonamide:
  • Example 40 Biological activity
  • Human GST-MEK1 and the constitutively 5 active allele GST-MEK1 CA can be subcloned into the yeast expression vector pGEM4Z (Promega, Madison, WI) from the wild type human MEK1 cDNA.
  • GST-MEK1CA was expressed in Escherichia coli and can be partially purified using Glutathione Sepharose 4B affinity resin (Amersham Pharmacia Biotech, Piscataway, NJ).
  • the ERK2 allele can be subcloned from MAPK2/Erk2 cDNA (wild type) in 10 pUSEamp (Upstate Biotechnology, Inc., Waltham, MA) into the vector pET21a (Novagen, Madison, WI) resulting in an N-terminal histidine-tagged mouse ERK2 allele.
  • ERK2 can be expressed and purified to homogeneity [Zhang, 1993 #33].
  • Myelin basic protein (MBP) can be purchased from Gibco BRL (Rockville, MD). EasyTides adenosine 5'-triphosphate (ATP) ([ ⁇ -33P]) (NEN Perkin Elmer, Wellesley, MA) is the source of radiolabel for all kinase reactions.
  • Activated Raf-1 truncated
  • activated MAPKinase 2/ERK2 can be purchased from Upstate, Inc. (Lake Placid, NY). 4-20% Criterion Precast gels can be purchased from Bio-Rad (Hercules, CA).
  • enzymatic activity Compounds are diluted from dimethylsulfoxide (DMSO) stocks into 1xHMNDE (20 mM HEPES pH 7.2, 1 mM MgCl2, 100 mM NaCl, 1.25 mM DTT, 0.2 mM EDTA).
  • a typical 25-microliter assay contained 0.002 nanomoles MEK1 CA , 0.02 nanomoles ERK2, 0.25 nanomoles MBP, 0.25 nanomoles unlabeled ATP, and 0.1 ⁇ Ci [ ⁇ 33P] ATP.
  • the screening assay essentially comprises four additions. Five ⁇ l of diluted compound are dispensed to 96-well assay plates.
  • a truncated version of MEK that requires activation by Raf kinase can be used.
  • Effects of compounds in the cell can be determined by Western blotting for phosphorylated ERK.
  • MDA-MB-231 breast cancer cells are plated in a 48 well plate at 20,000 cells per well and are grown in a 37° humidified CO 2 incubator. The following day, the growth media (DMEM + 10% fetal bovine serum) is removed and replaced with starve media (DMEM + 0.1% fetal bovine serum). Cells are incubated in the starve media for sixteen hours and then treated with a range of compound concentrations for thirty minutes.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Diabetes (AREA)
  • Hematology (AREA)
  • Dermatology (AREA)
  • Rheumatology (AREA)
  • Emergency Medicine (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Ophthalmology & Optometry (AREA)
  • Pain & Pain Management (AREA)
  • Urology & Nephrology (AREA)
  • Vascular Medicine (AREA)
  • Cardiology (AREA)
  • Endocrinology (AREA)
  • Obesity (AREA)
  • Oncology (AREA)
  • Immunology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Quinoline Compounds (AREA)
  • Heterocyclic Carbon Compounds Containing A Hetero Ring Having Nitrogen And Oxygen As The Only Ring Hetero Atoms (AREA)
  • Nitrogen And Oxygen Or Sulfur-Condensed Heterocyclic Ring Systems (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Description

    BACKGROUND OF THE INVENTION
  • Cell signaling pathways play an important role in cell growth, proliferation and differentiation. In normal cell growth, growth factors, through receptor activation (e.g. PDGF or EGF), activate MAP kinase pathways. The Ras/Raf kinase pathway is one of the most important and most well understood MAP kinase pathways involved in normal and uncontrolled cell growth. Active GTP-bound Ras leads to the activation of a cascade of serine/threonine kinases. One of the several groups of kinases known to require GTP-bound Ras for their activation is the Raf family. Upon activation Raf phosphorylates "mitogen-activated ERK activating kinases" (MEK) - 1 and MEK2.
  • Some cancers contain mutations which result in the continuous activation of this pathway due to continuous production of growth factors. Other mutations can lead to defects in the deactivation of the activated GTP-bound Ras complex, again resulting in activation of the MAP kinase pathway. Mutated, oncogenic forms of Ras are found in 50% of colon and >90% pancreatic cancers as well as many others types of cancers. Recently, bRaf mutations have been identified in more than 60% of malignant melanoma.
  • These mutations in bRaf result in a constitutively active MAP kinase cascade. Studies of primary tumor samples and cell lines have also shown constitutive or over activation of the MAP kinase pathway in cancers of pancreas, colon, lung, ovary and kidney. Hence, there is a strong correlation between cancers and an overactive MAP kinase pathway resulting from genetic mutations.
  • As constitutive or over activation of MAP kinase cascade plays a pivotal role in cell proliferation and differentiation, inhibition of this pathway is believed to be beneficial in hyperproliferative diseases. MEK is a key player in this pathway as it is downstream of Ras and Raf. Additionally, it is an attractive therapeutic target because the only known substrates for MEK phosphorylation are the MAP kinases, ERK1 and 2.
  • Inhibition of MEK has been shown to have potential therapeutic benefit in several studies. For example, small molecule MEK inhibitors have been shown to inhibit human tumor growth in nude mouse xenografts, block static allodynia in animals and inhibit growth of acute myeloid leukemia cells.
  • Thus, MEK1 and MEK2 are validated and accepted targets for anti-proliferative therapies, even when the oncogenic mutation does not affect MEK structure or expression.
  • The MEK cascade has also been implicated in inflammatory diseases and disorders. This includes both acute and chronic inflammation disorders. Examples of such disorders are allergic contact dermatitis, rheumatoid arthritis, osteoarthritis, inflammatory bowel diseases, chronic obstructive pulmonary disorder, psoriasis, multiple sclerosis, asthma, diseases and disorders related to diabetic complications, and inflammatory complications of the cardiovascular system such as acute coronary syndrome. Among inflammatory bowel diseases are Crohn's disease and ulcerative colitis.
  • PCT application published under WO 2004/083167 A (Sankyo Co ), relates to sulfamide derivatives and medicinal compositions thereof.
  • Li et al. in J. Med. CHem., 1995, 38(25), pp. 4897 to 4905, describe cyclooxygenase-2 inhibitors, in particular the synthesis and pharmacological activities of 5-Methanesulfonamido-1-indanone derivatives.
  • European patent application published under EP 0 679 641 A (Eisai Co Ltd) relates to sulfonamide derivatives and sulfonic ester derivatives having anti-tumour activity.
  • SUMMARY OF THE INVENTION
  • The invention provides compounds and methods of utilizing such compounds. The compounds and the pharmaceutically acceptable salts, esters, hydrates and solvates thereof of this invention are useful, e.g. in the treatment of diseases, e.g. hyperproliferative diseases. In one aspect, the invention provides compounds, including pharmaceutically acceptable salts, esters, hydrates and solvates thereof having the general formula I:
    Figure imgb0001
    where G is R1a, R1b, R1c, R1d, R1e, Ar1, Ar2 or Ar3; Ro, R1 and R2 are independently selected from H, halogen, cyano, cyanomethyl, nitro, difluoromethoxy, difluoromethoxy, trifluoromethyl, azido, CO2R5, OR5, -O-(CO)-R5, -O-C(O)-N(R5)2, -NR5C(O)NR6R7, -SR5, NHC(O)R5, -NHSO2R5, SO2N(R5)2, C1-C6 alkyl, C1-C4 alkoxy, C3-C6 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, aryl, alkylaryl, arylalkyl, heterocyclic, said alkyl, cycloalkyl, alkenyl, aryl, alkylaryl, arylalkyl, heterocyclic and alkynyl groups optionally substituted with 1-3 substituents selected independently from halogen, OH, CN, cyanomethyl, nitro, phenyl, difluoromethoxy, difluoromethoxy, and trifluoromethyl, and said C1-C6 alkyl and C1-C4 alkoxy groups also optionally substituted with OCH3 or OCH2CH3, wherein each R5 is selected from H, lower alkyl, substituted lower alkyl, aryl, or substituted aryl, and NR5R6, wherein each R6 and R7 are independently selected from hydrogen or lower alkyl; X is F, Br, I, Cl or methyl; Y is I, Br, Cl, CF3, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, cyclopropyl, phenyl, pyridyl, pyrazolyl, OMe, OEt, or SMe, where all said methyl, ethyl, C1-C3 alkyl, and cyclopropyl groups of X and Y are optionally substituted with OH, all said phenyl, pyridyl, pyrazolyl groups of Y are optionally substituted with halogen, acetyl, methyl, and trifluoromethyl, and all said methyl groups of X and Y are optionally substituted with one, two, or three F atoms; Z is H, F, Br, I, or Cl; and A, D, J, L, Q, U are independently selected from C, - NH, N, O, and -N(CH3)-.
  • R1a is methyl, optionally substituted with 1-3 fluorine atoms or 1-3 chlorine atoms, or with OH, cyclopropoxy, or C1- C4 alkoxy, where the C1- C4 alkyl moieties of said C1- C4 alkoxy groups are optionally substituted with one hydroxy or methoxy group, and where all C2- C4 alkyl groups within said C1- C4 alkoxy are optionally further substituted with a second OH group.
  • R1b is CH(CH3)-C1-3 alkyl or C3-C6 cycloalkyl, said methyl, alkyl, and cycloalkyl groups optionally substituted with 1-3 substituents selected independently from F, Cl, Br, I, OH, C1-C4 alkoxy, and CN.
  • R1c is (CH2)nOmR', where m is 0 or 1; where, when m is 1, n is 2 or 3, and when m is 0, n is 1 or 2; and where R' is C1-C6 alkyl, optionally substituted with 1-3 substituents selected independently from F, Cl, OH, OCH3, OCH2CH3, and C3-C6 cycloalkyl;
  • R1d is C(A')(A")(B)- where B, A', and A" are, independently, H or C1-4 alkyl, optionally substituted with one or two OH groups or halogen atoms, or A' and A", together with the carbon atom to which they are attached, form a 3- to 6- member saturated ring, said ring optionally containing one or two heteroatoms selected, independently, from O, N, and S and optionally substituted with one or two groups selected independently from methyl, ethyl, and halo.
  • R1e is benzyl or 2-phenyl ethyl, in which the phenyl group is optionally substituted
    Figure imgb0002
    where q is 1 or 2, Rs, R9 and R10 are, independently, H, F, Cl, Br, I, CH3, CH2F, CHF2, CF3, OCH3, OCH2F, OCHF2, OCF3, ethyl, n-propyl, isopropyl, cyclopropyl, isobutyl, sec-butyl, tert-butyl, and methylsulfonyl, and R10 may also be nitro, acetamido, amidinyl, cyano, carbamoyl, methylcarbamoyl, dimethylcarbamoyl, 1,3,4-oxadiazol-2-yl, 5-methyl-1,3,4-5 oxadiazolyl, 1,3,4-thiadiazolyl, 5-methyl-1,3,4-thiadiazol-1H-tetrazolyl, N-morpholinyl carbonylamino, N-morpholinylsulfonyl, and N-pyrrolidinylcarbonylamino; R11 and R12 are, independently, H, F, Cl, or methyl.
  • Ar1 is
    Figure imgb0003
    where W and V are, independently, N, CR2 or CR3; R3, Rs, R8 and R10 are, independently, H, F, Cl, Br, I, CH3, CH2F, CHF2, CF3, OCH3, OCH2F, OCHF2, OCF3, ethyl, n-propyl, isopropyl, cyclopropyl, isobutyl, sec-butyl, tert-butyl, and methylsulfonyl, and R10 may also be nitro, acetamido, amidinyl, cyano, carbamoyl, methylcarbamoyl, dimethylcarbamoyl, 1,3,4-oxadiazol-2-yl, 5-methyl-1,3,4-oxadiazol, 1,3,4-thiadiazol, 5-methyl-1,3,4-thiadiazol 1H-tetrazolyl, N-morpholinylcarbonylamino, N-morpholinylsulfonyl and Npyrrolidinylcarbonylamino; R11 and R12 are, independently, H, F, Cl or methyl.
  • Ar2 is
    Figure imgb0004
    where the dashed line represents a double bond which may be located formally either between V and the carbon between W and V, or between W and the carbon between W and V; where W is -S-, -O- or -N = and where, when W is -O- or -S-, V is -CH=, -CCl= or -N =; and when W is -N =, V is CH=, or -NCH3-; R13 and R14 are, independently, H, methoxycarbonyl, methylcarbamoyl, acetamido, acetyl, methyl, ethyl, trifluoromethyl, or halogen.
  • Ar3 is
    Figure imgb0005
    where W is -NH-, -NCH3- or -O-; and R13 and R8 are, independently, H, F, Cl, or methyl.
  • Such compounds are inhibitors of MEK and are useful in treatment of cancer and other hyperproliferative diseases.
  • This invention is also directed to pharmaceutical compositions comprising pharmaceutically effective amounts of a compound of formula I or a pharmaceutically acceptable salt, ester, solvate or hydrate thereof, and a pharmaceutically acceptable carrier. Such compositions may contain adjuvants, excipients, preservatives, agents for delaying absorption, fillers, binders, adsorbents, buffers, disintegrating agents, solubilizing agents, other carriers, and other inert ingredients. Methods of formulation of such compositions are well-known in the art.
  • The invention is also directed to use of a compound of formula I, or a pharmaceutically acceptable salt, ester, solvate or hydrate thereof, for the preparation of a medicament for the treatment of a hyperproliferative disease.
  • The invention is also directed to use of a compound of formula I, or a pharmaceutically acceptable salt, ester, solvate or hydrate thereof, for the preparation of a medicament for the treatment of a disorder or condition modulated by the MEK cascade. The appropriate dosage for a particular patient can be determined, according to known methods, by those skilled in the art.
  • DETAILED DESCRIPTION OF THE INVENTION
  • Reference will now be made in detail to particularly preferred embodiments of the invention. Examples of the preferred embodiments are illustrated in the following Examples section.
  • Unless defined otherwise, all technical and scientific terms used herein have the same meaning as is commonly understood by one of skill in the art to which this invention belongs.
  • Novel compounds encompassed by the instant invention include those described by the general formulas I and II set forth above, and the pharmaceutically acceptable salts, esters,solvates or hydrates thereof.
  • As used herein a "pharmaceutically acceptable salt" includes salts that retain the biological effectiveness of the free acids and bases of the specified compound and that are not biologically or otherwise undesirable. A compound of this invention may possess acidic or basic groups and therefore may react with any of a number of inorganic or organic bases, and inorganic and organic acids, to form a pharmaceutically acceptable salt. Examples of pharmaceutically acceptable salts include those salts prepared by reaction of the compounds of this invention with a mineral or organic acid or an inorganic base, such salts including sulfates, pyrosulfates, bisulfates, sulfites, bisulfites, phosphates, monohydrogenphosphates, dihydrogenphosphates, metaphosphates, pyrophosphates, chlorides, bromides, iodides, acetates, propionates, decanoates, caprylates, acrylates, formates, isobutyrates, caproates, heptanoates, propiolates, oxalates, malonates, succinates, suberates, sebacates, fumarates, maleates, butyn-1,4-dioates, hexyne-1,6-dioates, benzoates, chlorobenzoates, methylbenzoates, dinitrobenzoates, hydroxybenzoates, methoxybenzoates, phthalates, sulfonates, xylenesulfonates, phenylacetates, phenylpropionates, phenylbutyrates, citrates, lactates, .γ-hydroxybutyrates, glycollates, tartrates, methanesulfonates, propanesulfonates, naphthalene-1-sulfonates, naphthalene-3-sulfonates, and mandelates.
  • The invention provides compounds, including pharmaceutically acceptable saltsand solvates thereof having the formula :
    Figure imgb0006
    Figure imgb0007
    Figure imgb0008
    Figure imgb0009
    wherein
    • G is R1a, R1b, R1c, R1d, R1e, Ar1, Ar2 or Ar3;
    • Ro, R1 and R2 are independently selected from H, halogen, cyano, cyanomethyl, nitro, difluoromethoxy, difluoromethoxy, trifluoromethyl, azido, C1-C6 alkyl, C1-C4 alkoxy, C3-C6 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl; wherein
      • said C1-C6 alkyl, C3-C6 cycloalkyl, C2-C6 alkenyl and C2-C6 alkynyl groups are optionally substituted with 1-3 substituents selected independently from halogen, OH, CN, cyanomethyl, nitro, phenyl, difluoromethoxy, difluoromethoxy, and trifluoromethyl;
      • said C1-C6 alkyl and C1-C4 alkoxy groups are optionally substituted with OCH3 or OCH2CH3;
    • X is F, Cl or methyl;
    • Y is I, Br, Cl, CF3, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, cyclopropyl, phenyl, pyridyl, pyrazolyl, OMe, OEt, or SMe, wherein
      • all said methyl, ethyl, C1-C3 alkyl, and cyclopropyl groups of X and Y are optionally substituted with OH;
      • all said phenyl, pyridyl, pyrazolyl groups of Y are optionally substituted with halogen, acetyl, methyl, and trifluoromethyl; and
      • all said methyl groups of X and Y are optionally substituted with one, two, or three F atoms;
    • Z is H, methyl, Cl or F;
      and wherein
    • R1a is methyl, cyclopropoxy or C1- C4 alkoxy; wherein
      • the methyl is optionally substituted with OH, 1-3 fluorine atoms or 1-3 chlorine atoms;
      • the C1- C4 alkyl moieties of said C1- C4 alkoxy are optionally substituted with one hydroxy or methoxy group; and
      • all C2- C4 alkyl groups within said C1- C4 alkoxy are optionally further substituted with a second OH group;
    • R1b is CH(CH3)-C1-3 alkyl or C3-C6 cycloalkyl, said CH3, alkyl, and cycloalkyl groups optionally substituted with 1-3 substituents selected independently from F, Cl, Br, I, OH, C1-C4 alkoxy and CN.
    • R1c is (CH2)nOmR', where
      • m is 0 or 1; wherein
        • when m is 1, n is 2 or 3, and
        • when m is 0, n is 1 or 2;
      • R' is C1-C6 alkyl, optionally substituted with 1-3 substituents selected independently from F, Cl, OH, OCH3, OCH2CH3, and C3-C6 cycloalkyl;
    • R1d is C(A')(A")(B)- wherein
      • B, A', and A" are, independently, H or C1-4 alkyl, optionally substituted with one or two OH groups or halogen atoms, or
      • A' and A", together with the carbon atom to which they are attached, form a 3- to 6- member saturated ring, said ring optionally containing one or two heteroatoms selected, independently, from O, N, and S and optionally substituted with one or two groups selected independently from methyl, ethyl, and halo;
    • R1e is benzyl or 2-phenyl ethyl, in which the phenyl group is optionally substituted
      Figure imgb0010
      where
      • q is 1 or 2;
      • R8 and R9 are, independently, H, F, Cl, Br, CH3, CH2F, CHF2, CF3, OCH3, OCH2F, OCHF2, OCF3, ethyl, n-propyl, isopropyl, cyclopropyl, isobutyl, sec-butyl, tert-butyl, and methylsulfonyl;
      • R10 is H, F, Cl, Br, CH3, CH2F, CHF2, CF3, OCH3, OCH2F, OCHF2, OCF3, ethyl, n-propyl, isopropyl, cyclopropyl, isobutyl, sec-butyl, tert-butyl, and methylsulfonyl, nitro, acetamido, amidinyl, cyano, carbamoyl, methylcarbamoyl, dimethylcarbamoyl, 1,3,4-oxadiazol-3-yl, 5-methyl-1,3,4-5 oxadiazolyl, 1,3,4-thiadiazolyl, 5-methyl-1,3,4-thiadiazol-1H-tetrazolyl, N-morpholinyl carbonylamino, N-morpholinylsulfonyl or N-pyrrolidinylcarbonylamino;
      • R11 and R12 are, independently, H, F, Cl, or methyl;
    • Ar1 is
      Figure imgb0011
      where
      • W and V are, independently, N, CR1 or CR2;
      • Rs, R9 and R10 are, independently, H, F, Cl, Br, CH3, CH2F, CHF2, CF3, OCH3, OCH2F, OCHF2, OCF3, ethyl, n-propyl, isopropyl, cyclopropyl, isobutyl, sec-butyl, tert-butyl, and methylsulfonyl, and R10 may also be nitro, acetamido, amidinyl, cyano, carbamoyl, methylcarbamoyl, dimethylcarbamoyl, 1,3,4-oxadiazol-3-yl, 5-methyl-1,3,4-oxadiazol, 1,3,4-thiadiazol, 5-methyl-1,3,4-thiadiazol 1H-tetrazolyl, N-morpholinylcarbonylamino, N-morpholinylsulfonyl and N-pyrrolidinylcarbonylamino;
      • R11 and R12 are, independently, H, F, Cl or methyl;
    • Ar2 is
      Figure imgb0012
      where
      • the dashed line represents a double bond which may be located formally either between V and the carbon between W and V, or between W and the carbon between W and V;
      • W is -S-, -O- or -N =, wherein
        • when W is -O- or -S-, V is -CH=, -CCl= or -N =; and
        • when W is -N =, V is CH= or -NCH3-;
      • R13 and R14 are, independently, H, methoxycarbonyl, methylcarbamoyl, acetamido, acetyl, methyl, ethyl, trifluoromethyl or halogen;
    • Ar3 is
      Figure imgb0013
      where
      • W is -NH-, -NCH3- or -O-; and
      • R13 and R14 are, independently, H, F, Cl, or methyl.
  • In a first aspect, the present invention provides compounds of formula (I-A), (I-B), (I-C) or (I-D), where
    X is F, Cl, or CH3;
    Y is I, Br, Cl, CF3, or C1-C3 alkyl; and
    Z is H or F.
  • In a second aspect, the present invention provides compounds of formula (I-A), (I-B), (I-C) or (I-D), where
    Ro is F, Cl, C1-C4 alkyl or C1-C4 alkoxy, said C1-C4 alkyl group and the C1-C4 alkyl moiety of said C1-C4 alkoxy group optionally substituted with F, Cl, OCH3, or OCH2CH3.
  • In an embodiment of the first aspect, the present invention provides compounds of formula (I-A), (I-B), (I-C) or (I-D), where
    Ro is H, F, Cl, C1-C4 alkyl, methoxy, ethoxy, or 2-methoxy-ethoxy.
  • In a third aspect, the present invention provides compounds of formula (I-A), (I-B9, (I-C) or (I-D), where G is R1d.
  • In a first embodiment of the third aspect, the present invention provides compounds of formula (I-A), (I-B), (I-C) or (I-D), where
    Ro is either :
    • fluoro, chloro, methyl, ethyl, propyl, isopropyl, sec-butyl, iso-butyl, tert-butyl, cyclopropyl, cyclobutyl, fluoromethyl, methoxy, fluoromethoxy, methylamino or dimethylamino; when
      X is F, Cl, CH3, or mono-, di- or trifluoromethyl;
      or :
    • F, Cl, methyl, ethyl, methoxy, ethoxy, or 2-methoxy-ethoxy; when
      X is F, Cl, or CH3;
      or :
    • H; when
      X is F, Cl, CH3, or mono-, di- or trifluoromethyl;
    and
    Y is I, Br, Cl, or mono-, di- or tri- fluoromethyl; and
    Z is H or F.
  • In a second embodiment of the third aspect, the present invention provides compounds of formula (I-A), (I-B), (I-C) or (I-D), where C(A')(A") is C1-C6 cycloalkyl.
  • In a first variant of the second embodiment of the third aspect, the present invention provides compounds of formula (I-A), (I-B), (I-C) or (I-D), where B is H.
  • In an alternative of the first variant of the second embodiment of the third aspect, the present invention provides compounds of formula (I-A), (I-B), (I-C) or (I-D), where C(A')(A") is cyclopropyl.
  • In a second variant of the second embodiment of the third aspect, the present invention provides compounds of formula (I-A), (I-B), (I-C) or (I-D), where B is methyl, optionally substituted with one OH group, or C2-C4 alkyl, optionally substituted with one or two OH groups.
  • In an alternative of the second variant of the second embodiment of the third aspect, the present invention provides compounds of formula (I-A), (I-B), (I-C) or (I-D), where C(A')(A") is cyclopropyl.
  • In another alternative of the second variant of the second embodiment of the third aspect, the present invention provides compounds of formula (I-A), (I-B), (I-C) or (I-D), where B is methyl, ethyl, 2-hydroxyethyl, n-propyl, 3-5 hydroxypropyl, 2,3-dihydroxypropyl, 3,4-dihydroxybutyl, isopropyl, 1-methyl-2-hydroxyethyl, n-butyl, sec-butyl, isobutyl, or 2-hydroxymethyl-3-hydroxy propyl.
  • In another alternative of the second variant of the second embodiment of the third aspect, the present invention provides compounds of formula (I-A), (I-B), (I-C) or (I-D), where B is 2,3-dihydroxypropyl or 3,4-dihydroxybutyl.
  • In another alternative of the second variant of the second embodiment of the third aspect, the present invention provides compounds of formula (I-A), (I-B), (I-C) or (I-D), in which the chiral carbon in B is in the R configuration.
  • Compounds of the following formulae are exemplary compounds of formula I :
    Figure imgb0014
    Figure imgb0015
    Figure imgb0016
    Figure imgb0017
    Figure imgb0018
    Figure imgb0019
    Figure imgb0020
    Figure imgb0021
    Figure imgb0022
    Figure imgb0023
    Figure imgb0024
    Figure imgb0025
    Figure imgb0026
    Figure imgb0027
    Figure imgb0028
    Figure imgb0029
    Figure imgb0030
    Figure imgb0031
    and
    Figure imgb0032
  • Certain compounds of the present invention may have asymmetric centers and therefore exist in different enantiomeric forms. All optical isomers and stereoisomers of the compounds of the present invention, and mixtures thereof, are considered to be within the scope of the invention. With respect to the compounds of the present invention, the invention includes the use of a racemate, one or more enantiomeric forms, one or more diastereomeric forms, or mixtures thereof. The compounds of the present invention may also exist as tautomers. This invention relates to the use of all such tautomers and mixtures thereof. The compounds of the present invention may also exist in different polymorphic states. This invention relates to the use of all such polymorphic states and mixtures thereof.
  • The subject invention also includes isotopically-labeled compounds, which are identical to those recited in the present invention, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Examples of isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, sulfur, fluorine and chloride, such as 2H, 3H, 13C, 14C, 15N, 180, 17O, 31P, 32P, 35S, 18F, and 36Cl, respectively. Compounds of the present invention, pharmaceutically acceptable salt, ester, solvate or hydrate thereof which contain the aforementioned isotopes and/or other isotopes of other atoms are within the scope of this invention. Certain isotopically-labeled compounds of the present invention, for example those into which radioactive isotopes such as 3H and 14C are incorporated, are useful in drug and/or substrate tissue distribution assays.
  • Tritiated, i. e., 3H and carbon-14, i. e., 14C, isotopes are particularly preferred for their ease of preparation and detectability. Further, substitution with heavier isotopes such as deuterium, i. e., 2H, can afford certain therapeutic advantages resulting from greater metabolic stability, for example increased in vivo half-life or reduced dosage requirements and, hence, may be preferred in some circumstances. Isotopically labeled compound of the present invention, pharmaceutically acceptable salt, ester, solvate or hydrate thereof can generally be prepared by carrying out procedures disclosed in the Schemes and/or in the Examples and Preparations below, by substituting a readily available isotopically labeled reagent for a non-isotopically labeled reagent.
  • It is to be understood that in instances where two or more radicals are used in succession to define a substituent attached to a structure, the first named radical is considered to be terminal and the last named radical is considered to be attached to the structure in question. Thus, for example, the radical arylalkyl is attached to the structure in question by the alkyl group.
  • The invention also relates to pharmaceutical compositions. In some embodiments the invention provides pharmaceutical compositions for the treatment of disorders such as hyperproliferative disorder in a mammal. In some embodiment, the treatment of said disorders comprises a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt, ester, solvate or hydrate thereof, and a pharmaceutically acceptable carrier. In some embodiments, said pharmaceutical composition is for the treatment of cancer such as acute myeloid leukemia, thymus, brain, lung, squamous cell, skin, eye, retinoblastoma, intraocular melanoma, oral cavity and oropharyngeal, bladder, gastric, stomach, pancreatic, bladder, breast, cervical, head, neck, renal, kidney, liver, ovarian, prostate, colorectal, esophageal, testicular, gynecological, thyroid, CNS, PNS, AIDS related AIDS-Related (e.g. Lymphoma and Kaposi's Sarcoma) or Viral-Induced cancer. In some embodiments, said pharmaceutical composition is for the treatment of a non-cancerous hyperproliferative disorder such as benign hyperplasia of the skin (e. g., psoriasis), restenosis, or prostate (e. g., benign prostatic hypertrophy (BPH)).
  • The invention also relates to a pharmaceutical composition for the treatment of pancreatitis or kidney disease (including proliferative glomerulonephritis and diabetes- induced renal disease) or pain in a mammal which comprises a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt, ester, solvate or hydrate thereof, and a pharmaceutically acceptable carrier.
  • The invention also relates to a pharmaceutical composition for the prevention of blastocyte implantation in a mammal which comprises a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt, ester, solvate or hydrate thereof, and a pharmaceutically acceptable carrier.
  • The invention also relates to a pharmaceutical composition for treating a disease related to vasculogenesis or angiogenesis in a mammal which comprises a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt, ester, solvate or hydrate thereof, and a pharmaceutically acceptable carrier. In some embodiments, said pharmaceutical composition is for treating a disease selected from the group consisting of tumor angiogenesis, chronic inflammatory disease such as rheumatoid arthritis, inflammatory bowel disease, atherosclerosis, skin diseases such as psoriasis, eczema, and scleroderma, diabetes, diabetic retinopathy, retinopathy of prematurity, age-related macular degeneration, hemangioma, glioma, melanoma, Kaposi's sarcoma and ovarian, breast, lung, pancreatic, prostate, colon and epidermoid cancer.
  • The invention also relates to use of a compound of the present invention, or a pharmaceutically acceptable salt, ester, solvate or hydrate thereof for the preparation of a medicament for treating a hyperproliferative disorder. In some embodiments, said use is in the treatment of cancer such as acute myeloid leukemia, thymus, brain, lung, squamous cell, skin, eye, retinoblastoma, intraocular melanoma, oral cavity and oropharyngeal, bladder, gastric, stomach, pancreatic, bladder, breast, cervical, head, neck, renal, kidney, liver, ovarian, prostate, colorectal, esophageal, testicular, gynecological, thyroid, CNS, PNS, AIDS related AIDS-Related (e.g. Lymphoma and Kaposi's Sarcoma) or Viral-Induced cancer. In some embodiments, said use is in the treatment of a non-cancerous hyperproliferative disorder such as benign hyperplasia of the skin (e. g., psoriasis), restenosis, or prostate (e. g., benign prostatic hypertrophy (BPH)).
  • The invention also relates to use of a compound of the present invention, or a pharmaceutically acceptable salt, ester, solvate or hydrate thereof, in combination with an anti-tumor agent for the preparation of a medicament for the treatment of a hyperproliferative disorder. In some embodiments, the anti-tumor agent is selected from the group consisting of mitotic inhibitors, alkylating agents, anti- metabolites, intercalating antibiotics, growth factor inhibitors, cell cycle inhibitors, enzyme inhibitors, topoisomerase inhibitors, biological response modifiers, anti- hormones, angiogenesis inhibitors, and anti-androgens.
  • The invention also relates to use of a compound of the present invention, or a pharmaceutically acceptable salt, ester, solvate or hydrate thereof, for the preparation of a medicament for the treatment of diseases related to vasculogenesis or angiogenesis. In some embodiments, said use is in treating a disease selected from the group consisting of tumor angiogenesis, chronic inflammatory disease such as rheumatoid arthritis, atherosclerosis, inflammatory bowel disease, skin diseases such as psoriasis, eczema, and scleroderma, diabetes, diabetic retinopathy, retinopathy of prematurity, age-related macular degeneration, hemangioma, glioma, melanoma, Kaposi's sarcoma and ovarian, breast, lung, pancreatic, prostate, colon and epidermoid cancer.
  • Patients that can be treated with compounds of the present invention, or pharmaceutically acceptable salt, ester, solvate or hydrate of said compounds, include, for example, patients that have been diagnosed as having psoriasis; restenosis; atherosclerosis; BPH; breast cancer such as a ductal carcinoma in duct tissue in a mammary gland, medullary carcinomas, colloid carcinomas, tubular carcinomas, and inflammatory breast cancer; ovarian cancer, including epithelial ovarian tumors such as adenocarcinoma in the ovary and an adenocarcinoma that has migrated from the ovary into the abdominal cavity; uterine cancer; cervical cancer such as adenocarcinoma in the cervix epithelial including squamous cell carcinoma and adenocarcinomas; prostate cancer, such as a prostate cancer selected from the following: an adenocarcinoma or an adenocarinoma that has migrated to the bone; pancreatic cancer such as epitheliod carcinoma in the pancreatic duct tissue and an adenocarcinoma in a pancreatic duct; bladder cancer such as a transitional cell carcinoma in urinary bladder, urothelial carcinomas (transitional cell carcinomas), tumors in the urothelial cells that line the bladder, squamous cell carcinomas, adenocarcinomas, and small cell cancers; leukemia such as acute myeloid leukemia (AML), acute lymphocytic leukemia, chronic lymphocytic leukemia, chronic myeloid leukemia, hairy cell leukemia, myelodysplasia, and myeloproliferative disorders; bone cancer; lung cancer such as non-small cell lung cancer (NSCLC), which is divided into squamous cell carcinomas, adenocarcinomas, and large cell undifferentiated carcinomas, and small cell lung cancer; skin cancer such as basal cell carcinoma, melanoma, squamous cell carcinoma and actinic keratosis, which is a skin condition that sometimes develops into squamous cell carcinoma; eye retinoblastoma; cutaneous or intraocular (eye) melanoma; primary liver cancer (cancer that begins in the liver); kidney cancer; thyroid cancer such as papillary, follicular, medullary and anaplastic; AIDS-related lymphoma such as diffuse large B-cell lymphoma, B-cell immunoblastic lymphoma and small non-cleaved cell lymphoma; Kaposi's Sarcoma; viral-induced cancers including hepatitis B virus (HBV), hepatitis C virus (HCV), and hepatocellular carcinoma; human lymphotropic virus-type 1 (HTLV-1) and adult T-cell leukemia/lymphoma; and human papilloma virus (HPV) and cervical cancer; central nervous system cancers (CNS) such as primary brain tumor, which includes gliomas (astrocytoma, anaplastic astrocytoma, or glioblastoma multiforme), Oligodendroglioma, Ependymoma, Meningioma, Lymphoma, Schwannoma, and Medulloblastoma; peripheral nervous system (PNS) cancers such as acoustic neuromas and malignant peripheral nerve sheath tumor (MPNST) including neurofibromas and schwannomas, malignant fibrous cytoma, malignant fibrous histiocytoma, malignant meningioma, malignant mesothelioma, and malignant mixed Müllerian tumor; oral cavity and oropharyngeal cancer such as, hypopharyngeal cancer, laryngeal cancer, nasopharyngeal cancer, and oropharyngeal cancer; stomach cancer such as lymphomas, gastric stromal tumors, and carcinoid tumors; testicular cancer such as germ cell tumors (GCTs), which include seminomas and nonseminomas, and gonadal stromal tumors, which include Leydig cell tumors and Sertoli cell tumors; thymus cancer such as to thymomas, thymic carcinomas, Hodgkin disease, non-Hodgkin lymphomas carcinoids or carcinoid tumors; rectal cancer; and colon cancer
  • The invention further provides methods of modulating MEK activity by contacting MEK with an amount of a compound of the invention sufficient to modulate the activity of MEK. Modulate can be inhibiting or activating MEK activity. In some embodiments, the invention provides methods of inhibiting MEK activity by contacting MEK with an amount of a compound of the invention sufficient to inhibit the activity of MEK. In some embodiments, the invention provides methods of inhibiting MEK activity in a solution by contacting said solution with an amount of a compound of the invention sufficient to inhibit the activity of MEK in said solution. In some embodiments, the invention provides methods of inhibiting MEK activity in a cell by contacting said cell with an amount of a compound of the invention sufficient to inhibit the activity of MEK in said cell. In some embodiments, the invention provides methods of inhibiting MEK activity in a tissue by contacting said tissue with an amount of a compound of the invention sufficient to inhibit the activity of MEK in said tissue. In some embodiments, the invention provides methods of inhibiting MEK activity in an organism by contacting said organism with an amount of a compound of the invention sufficient to inhibit the activity of MEK in said organism. In some embodiments, the invention provides methods of inhibiting MEK activity in an animal by contacting said animal with an amount of a compound of the invention sufficient to inhibit the activity of MEK in said animal. In some embodiments, the invention provides methods of inhibiting MEK activity in a mammal by contacting said mammal with an amount of a compound of the invention sufficient to inhibit the activity of MEK in said mammal. In some embodiments, the invention provides methods of inhibiting MEK activity in a human by contacting said human with an amount of a compound of the invention sufficient to inhibit the activity of MEK in said human.
  • This invention also relates to a pharmaceutical composition for inhibiting abnormal cell growth in a mammal which comprises an amount of a compound of the present invention, or a pharmaceutically acceptable salt, ester, solvate or hydrate thereof, in combination with an amount of a chemotherapeutic, wherein the amounts of the compound, salt, ester, solvate or hydrate, and of the chemotherapeutic are together effective in inhibiting abnormal cell growth. Many chemotherapeutics are presently known in the art and can be used in combination with the compounds of the invention.
  • In some embodiments, the chemotherapeutic is selected from the group consisting of mitotic inhibitors, alkylating agents, anti-metabolites, intercalating antibiotics, growth factor inhibitors, cell cycle inhibitors, enzymes, topoisomerase inhibitors, biological response modifiers, anti-hormones, angiogenesis inhibitors, and anti-androgens.
  • This invention further relates to use of a compound of the present invention, or a pharmaceutically acceptable salt, ester, solvate or hydrate thereof, in combination with radiation therapy, in the treatment of inhibiting abnormal cell growth, wherein the amounts of the compound, salt, ester, solvate or hydrate, is in combination with the radiation therapy effective in inhibiting abnormal cell growth. Techniques for administering radiation therapy are known in the art, and these techniques can be used in the combination therapy described herein. The administration of the compound of the invention in this combination therapy can be determined as described herein.
  • Without be limiting to any theory, the compounds of the present invention can render abnormal cells more sensitive to treatment with radiation for purposes of killing and/or inhibiting the growth of such cells.
  • The invention also relates to use of a compound of the present invention, or a pharmaceutically acceptable salt, ester, solvate or hydrate thereof, or an isotopically-labeled derivative thereof, and an amount of one or more substances selected from anti-angiogenesis agents, signal transduction inhibitors, and antiproliferative agents, in the treatment of inhibiting abnormal cell growth.
  • Anti-angiogenesis agents, such as MAP-2 (matrix-metalloprotienase 2) inhibitors, MMP-9 (matrix-metalloprotienase 9) inhibitors, and COX-11 (cyclooxygenase 11) inhibitors, can be used in conjunction with a compound of the present invention and pharmaceutical compositions described herein. Examples of useful COX-II inhibitors include CELEBREX™ (alecoxib), valdecoxib, and rofecoxib. Examples of useful matrix metalloproteinase inhibitors are described in WO 96/33172 (published October 24,1996 ), WO 96/27583 (published March 7,1996 ), European Patent Application No. 97304971.1 (filed July 8,1997 ), European Patent Application No. 99308617.2 (filed October 29, 1999 ), WO 98/07697 (published February 26,1998 ), WO 98/03516 (published January 29,1998 ), WO 98/34918 (published August 13,1998 ), WO 98/34915 (published August 13,1998 ), WO 98/33768 (published August 6,1998 ), WO 98/30566 (published July 16, 1998 ), European Patent Publication 606,046 (published July 13,1994 ), European Patent Publication 931, 788 (published July 28,1999 ), WO 90/05719 (published May 31,1990 ), WO 99/53910 (published October 21,1999 ), WO 99/52889 (published October 21, 1999 ), WO 99/29667 (published June 17,1999 ), PCT International Application No. PCT/IB98/01113 (filed July 21,1998 ), European Patent Application No. 99302232.1 (filed March 25,1999 ), Great Britain Patent Application No. 9912961.1 (filed June 3, 1999 ), United States Provisional Application No. 60/148,464 (filed August 12,1999 ), United States Patent 5,863, 949 (issued January 26,1999 ), United States Patent 5,861, 510 (issued January 19,1999 ), and European Patent Publication 780,386 (published June 25, 1997 ). Preferred MMP-2 and MMP-9 inhibitors are those that have little or no activity inhibiting MMP-1. More preferred, are those that selectively inhibit MMP-2 and/or AMP-9 relative to the other matrix-metalloproteinases (i. e., MAP-1, MMP-3, MMP-4, MMP-5, MMP-6, MMP- 7, MMP-8, MMP-10, MMP-11, MMP-12, andMMP-13). Some specific examples of MMP inhibitors useful in the present invention are AG-3340, RO 32-3555, and RS 13-0830.
  • The term "treating" and its grammatical equivalents as used herein include achieving a therapeutic benefit and/or a prophylactic benefit. By therapeutic benefit is meant eradication or amelioration of the underlying disorder being treated. Also, a therapeutic benefit is achieved with the eradication or amelioration of one or more of the physiological symptoms associated with the underlying disorder such that an improvement is observed in the patient, notwithstanding that the patient may still be afflicted with the underlying disorder. For prophylactic benefit, the compositions may be administered to a patient at risk of developing a particular disease, or to a patient reporting one or more of the physiological symptoms of a disease, even though a diagnosis of this disease may not have been made.
  • In some embodiments the compounds of the invention will be co-administer with other agents as described above. The term "Co-administration," "administered in combination with," and their grammatical equivalents, as used herein, encompasses administration of two or more agents to an animal so that both agents and/or their metabolites are present in the animal at the same time. Co-administration includes simultaneous administration in separate compositions, administration at different times in separate compositions, or administration in a composition in which both agents are present. Thus, in some embodiments, the compounds of the invention and the other agent(s) are administered in a single composition. In some embodiments, compounds of the invention and the other agent(s) are admixed in the composition.
  • Synthetic Procedures
  • In another aspect, the invention provides methods for synthesizing the compounds described herein. In some embodiments, the compounds of this invention can be prepared by the methods described below. The procedures below are intended to illustrate those methods, and the examples given are intended to illustrate the scope of this invention. Neither the methods not the examples should be construed as limiting the invention in any way. The procedures below are explained in more detail in the examples section.
  • Included in the methods of synthesis of the invention is the preparation of compound of formula 1-17 (as labeled in scheme 1). Scheme 1 is outlined below:
    Figure imgb0033
  • Included in the methods of synthesis of the invention is the preparation of compounds of general formula 2-10 (as labeled in scheme 2). Scheme 2 is outlined below:
    Figure imgb0034
  • Included in the methods of synthesis of the invention is the preparation of compounds of general formula 3-13 (as labeled in scheme 3). Scheme 3 is outlined below:
    Figure imgb0035
  • Included in the methods of synthesis of the invention is the preparation of compounds of general formula 4-7 (as labeled in scheme 4). Scheme 4 is outlined below:
    Figure imgb0036
  • Included in the methods of synthesis of the invention is the preparation of compounds of general formula 5-14 (as labeled in scheme 5). Scheme 5 is outlined below:
    Figure imgb0037
  • Included in the methods of synthesis of the invention is the preparation of compounds of general formula 6-9 (as labeled in scheme 6). Scheme 6 is outlined below:
    Figure imgb0038
  • Included in the methods of synthesis of the invention is the preparation of compounds of general formula 7-13 (as labeled in scheme 7) Scheme 7 is outlined below:
    Figure imgb0039
  • Included in the methods of synthesis of the invention is the preparation of compounds of general formula 8-7 (as labeled in scheme 8) Scheme 8 is outlined below:
    Figure imgb0040
  • Included in the methods of synthesis of the invention is the preparation of compounds of general formula 9-7 (as labeled in scheme 9) Scheme 9 is outlined below:
    Figure imgb0041
  • Included in the methods of synthesis of the invention is the preparation of compounds of general formula 10-7 (as labeled in scheme 10). Scheme 10 is outlined below:
    Figure imgb0042
  • Included in the methods of synthesis of the invention is the preparation of compounds of general formula 11-14 (as labeled in scheme 11). Scheme 11 is outlined below:
    Figure imgb0043
  • Included in the methods of synthesis of the invention is the preparation of compounds of general formula 12-12 (as labeled in scheme 12). Scheme 12 is outlined below:
    Figure imgb0044
  • Included in the methods of synthesis of the invention is the preparation of compounds of general formula 13-10 (as labeled in scheme 13). Scheme 13 is outlined below:
    Figure imgb0045
  • Included in the methods of synthesis of the invention is the preparation of compounds of general formula 14-14 (as labeled in scheme 14). Scheme 14 is outlined below:
    Figure imgb0046
  • Included in the methods of synthesis of the invention is the preparation of compounds of general formula 15-9 and 15-121 (as labeled in scheme 15). Scheme 15 is outlined below:
    Figure imgb0047
  • Included in the methods of synthesis of the invention is the preparation of compounds of general formula 16-7 (as labeled in scheme 16). Scheme 16 is outlined below:
    Figure imgb0048
  • Included in the methods of synthesis of the invention is the preparation of compounds of general formula 17-7 (as labeled in scheme 17). Scheme 17 is outlined below:
    Figure imgb0049
  • Included in the methods of synthesis of the invention is the preparation of compounds of general formula 18-10 (as labeled in scheme 18). Scheme 18 is outlined below:
    Figure imgb0050
  • Included in the methods of synthesis of the invention is the preparation of compounds of general formula 19-7 (as labeled in scheme 19). Scheme 19 is outlined below:
    Figure imgb0051
  • The compounds of the present invention may have asymmetric carbon atoms. Diastereomeric mixtures can be separated into their individual diastereomers on the basis of their physical chemical differences by methods known to those skilled in the art, for example, by chromatography or fractional crystallization. Enantiomers can be separated by converting the enantiomer mixture into a diastereomeric mixture by reaction with an appropriate optically active compound (e. g. alcohol), separating the diastereomers and converting (e. g., hydrolyzing) the individual diastereomers to the corresponding pure enantiomers. All such isomers, including diastereomeric mixtures and pure enantiomers are considered as part of the invention.
  • The activity of the compounds of the present invention may be determined by the following procedure, as well as the procedure described in the examples below. N-terminal 6 His-tagged, constitutively active MEK1 (2-393) is expressed in E. coli and protein is purified by conventional methods (Ahn et al. Science 1994, 265, 966-970). The activity of MEK1 is assessed by measuring the incorporation of γ-33P-phosphate from γ -33P-ATP onto N-terminal His tagged ERK2, which is expressed in E. coli and is purified by conventional methods, in the presence of MEK1. The assay is carried out in 96-well polypropylene plate. The incubation mixture (100, µL) comprises of 25 mM Hepes, pH 7.4, 10 mM MgCl2, 5 mM β-glycerolphosphate, 100 µM Na-orthovanadate, 5 mM DTT, 5 nM MEK1, and 1 µM ERK2. Inhibitors are suspended in DMSO, and all reactions, including controls are performed at a final concentration of 1% DMSO. Reactions are initiated by the addition of 10 µM ATP (with 0.5 µCi γ-33P- ATP/well) and incubated at ambient temperature for 45 minutes. Equal volume of 25% TCA is added to stop the reaction and precipitate the proteins. Precipitated proteins are trapped onto glass fiber B filter plates, and excess labeled ATP washed off using a Tomtec MACH III harvestor. Plates are allowed to air-dry prior to adding 30 µL/well of Packard Microscint 20, and plates are counted using a Packard TopCount.
  • Administration of the compounds of the present invention (hereinafter the "active compound (s)") can be effected by any method that enables delivery of the compounds to the site of action. These methods include oral routes, intraduodenal routes, parenteral injection (including intravenous, subcutaneous, intramuscular, intravascular or infusion), topical, and rectal administration.
  • The amount of the active compound administered will be dependent on the mammal being treated, the severity of the disorder or condition, the rate of administration, the disposition of the compound and the discretion of the prescribing physician. However, an effective dosage is in the range of about 0.001 to about 100 mg per kg body weight per day, preferably about 1 to about 35 mg/kg/day, in single or divided doses. For a 70 kg human, this would amount to about 0.05 to 7 g/day, preferably about 0.05 to about 2.5 g/day. In some instances, dosage levels below the lower limit of the aforesaid range may be more than adequate, while in other cases still larger doses may be employed without causing any harmful side effect, e.g. by dividing such larger doses into several small doses for administration throughout the day.
  • The active compound may be applied as a sole therapy or may involve one or more other anti-tumor substances, for example those selected from, mitotic inhibitors, for example vinblastine; alkylating agents, for example cis-platin, carboplatin and cyclophosphamide; anti-metabolites, for example 5-fluorouracil, cytosine arabinside and hydroxyurea, or, for example, one of the preferred anti-metabolites disclosed in European Patent Application No. 239362 such as N- (5- [N- (3, 4-dihydro-2-methyl-4- oxoquinazolin-6-yhnethyl)-N-methylamino]-2-thenoyl)-L-glutamic acid; growth factor inhibitors; cell cycle inhibitors; intercalating antibiotics, for example adriamycin and bleomycin; enzymes, for example, interferon; and anti-hormones, for example anti- estrogens such as Nolvadex™ (tamoxifen) or, for example anti-androgens such as Casodex™ (4'-cyano-3-(4-fluorophenylsulphonyl)-2-hydroxy-2-methyl-3'- (trifluoromethyl) propionanilide). Such conjoint treatment may be achieved by way of the simultaneous, sequential or separate dosing of the individual components of treatment.
  • The pharmaceutical composition may, for example, be in a form suitable for oral administration as a tablet, capsule, pill, powder, sustained release formulations, solution, suspension, for parenteral injection as a sterile solution, suspension or emulsion, for topical administration as an ointment or cream or for rectal administration as a suppository. The pharmaceutical composition may be in unit dosage forms suitable for single administration of precise dosages. The pharmaceutical composition will include a conventional pharmaceutical carrier or excipient and a compound according to the invention as an active ingredient. In addition, it may include other medicinal or pharmaceutical agents, carriers, adjuvants, etc.
  • Exemplary parenteral administration forms include solutions or suspensions of active compounds in sterile aqueous solutions, for example, aqueous propylene glycol or dextrose solutions. Such dosage forms can be suitably buffered, if desired.
  • Suitable pharmaceutical carriers include inert diluents or fillers, water and various organic solvents. The pharmaceutical compositions may, if desired, contain additional ingredients such as flavorings, binders, excipients and the like. Thus for oral administration, tablets containing various excipients, such as citric acid may be employed together with various disintegrants such as starch, alginic acid and certain complex silicates and with binding agents such as sucrose, gelatin and acacia. Additionally, lubricating agents such as magnesium stearate, sodium lauryl sulfate and talc are often useful for tableting purposes. Solid compositions of a similar type may also be employed in soft and hard filled gelatin capsules. Preferred materials, therefore, include lactose or milk sugar and high molecular weight polyethylene glycols. When aqueous suspensions or elixirs are desired for oral administration the active compound therein may be combined with various sweetening or flavoring agents, coloring matters or dyes and, if desired, emulsifying agents or suspending agents, together with diluents such as water, ethanol, propylene glycol, glycerin, or combinations thereof.
  • Methods of preparing various pharmaceutical compositions with a specific amount of active compound are known, or will be apparent, to those skilled in this art. For examples, see Remington's Pharmaceutical Sciences, Mack Publishing Company, Ester, Pa., 18th Edition (1990).
  • In still another aspect, the present invention provides kits for the treatment of disorders, such as the ones described herein. These kits comprise a compound or compounds described herein in a container and, optionally, instructions teaching the use of the kit according to the various methods and approaches described herein. Such kits may also include information, such as scientific literature references, package insert materials, clinical trial results, and/or summaries of these and the like, which indicate or establish the activities and/or advantages of the composition, and/or which describe dosing, administration, side effects, drug interactions, or other information useful to the health care provider.. Such information may be based on the results of various studies, for example, studies using experimental animals involving in vivo models and studies based on human clinical trials. Kits described herein can be provided, marketed and/or promoted to health providers, including physicians, nurses, pharmacists, formulary officials, and the like. Kits may also, in some embodiments, be marketed directly to the consumer.
  • The examples and preparations provided below further illustrate and exemplify the compounds of the present invention and methods of preparing such compounds. It is to be understood that the scope of the present invention is not limited in any way by the scope of the following examples and preparations. In the following examples molecules with a single chiral center, unless otherwise noted, exist as a racemic mixture. Those molecules with two or more chiral centers, unless otherwise noted, exist as a racemic mixture of diastereomers. Single enantiomers/diastereomers may be obtained by methods known to those skilled in the art.
  • EXAMPLES Example 1: N-(4-fluoro-5-(2-fluoro-4-iodophenylamino)-1-methyl-1H-benzo[d]imidazol-6-yl)-1-(2-hydroxyethyl)cyclopropane-1-sulfonamide: Step A: 3,3,4-Trifluoro-5-nitrobenzoic acid:
  • Figure imgb0052
  • Fuming nitric acid (1.7 ml) is added dropwise to concentrated sulfuric acid (25 ml) while maintaining the temperature at 5 - 10 °C. 2,3,4-Trifluorobenzoic acid (5 g, 28 mmoles) is added in small portion to this solution while keeping the reaction temperature at 5 °C. After completion the reaction mixture is stirred at room temperature for an additional 2 hours and poured into ice. The mixture is extracted with ether (3 x 75 ml). The organic layers are combined, washed with brine, dried (MgSO4). The solvent is removed, and the crude product is recrystallized from hot chloroform to obtain the title compound.
  • Step B: 3,4-Difluoro-2-(2-fluoro-4-iodophenylamino)-5-nitrobenzoic acid:
  • Figure imgb0053
  • To a solution of 2-fluoro-4-iodoaniline (2.9 g, 11.8 mmoles) in 50 ml anhydrous THF at -60 °C, 40 ml of a 1M solution of LHMDS in THF (40 mmoles) is added dropwise. In a separate flask, 2,3,4-trifluoro-5-nitrobenzoic acid (5 g, 22.6 mmoles), previously dissolved in THF (50 ml), is treated, at -60 °C, with 25 ml of a 1M solution of LHMDS in THF (25 mmoles). Both solutions are stirred at -78 °C for 45 min and the second solution is transferred visa cannula to the first reaction mixture. After completion of the addition the resulting mixture is stirred under argon at room temperature for 15 hours. The reaction mixture is quenched with water, then 1N HCl is added (pH = 0-1) followed by brine (100 ml). The crude material is extracted with THF (3 x 100 ml), the organic layers are combined and dried (Na2SO4) and the solvent is removed to give the title compound.
  • Step C: Methyl 3,4-difluoro-2-(2-fluoro-4-iodophenylamino)-5-nitrobenzoate:
  • Figure imgb0054
  • To a solution of 3,4-difluoro-2-(2-fluoro-4-iodophenylamino)-6-nitrobenzoic acid (8.5 g, 194 mmoles) in acetone (50 ml) is added sodium bicarbonate (5 g, 58.2 mmoles). The reaction mixture is stirred at room temperature for 10 min, dimethylsulfate (4.5 ml, 44.6 mmoles) is added and the mixture is heated under reflux for 3 hours. The solvent is removed under reduced pressure. The yellow residue is triturated in hot methanol. The solid is isolated by filtration, washed with MeOH and dried under vacuum to obtain the title compound.
  • Step D: Methyl 4-(allylamino)-3-fluoro-2-(2-fluoro-4-iodophenylamino)-5-nitrobenzoate:
  • Figure imgb0055
  • A suspension of methyl 3,4-difluoro-2-(2-fluoro-4-iodophenylamino)-5-nitrobenzoate (3.9 g, 8.62 mmoles) and allylamine (3.5 ml, 43 mmoles) in a mixture of THF (10 ml), MeOH (10 ml) and H2O (2.5 ml) is stirred at room temperature for 30 min. The yellow suspension is filtered and washed with hexane to remove the excess of allylamine. The resulting solid is dried under vacuum to obtain the title compound.
  • Step E: Methyl 4-(allylamino)-5-amino-3-fluoro-2-(2-fluoro-4-iodophenylamino)benzoate:
  • Figure imgb0056
  • A suspension of methyl 4-(allylamino)-3-fluoro-2-(2-fluoro-4-iodophenylamino)-5-nitrobenzoate and ammonium chloride (4.42 g, 79 mmoles) in a mixture of MeOH (20 ml) and dioxane (20 ml) is heated until it became a clear solution. Iron powder (4.42 g, 79 mmoles) is added and the resulting mixture is heated under reflux overnight. Water is added and the reaction mixture is filtered through celite. The solvent is removed under reduced pressure and the crude material is purified by flash chromatography to give the title compound.
  • Step F: Methyl 1-allyl-7-fluoro-6-(2-fluoro-4-iodophenylamino)-1H-benzo[d]imidazole-5-carboxylate:
  • Figure imgb0057
  • A stirred suspension of methyl 4-(allylamino)-5-amino-3-fluoro-2-(2-fluoro-4-iodophenylamino)benzoate (3.4 g, 7.4 mmoles)) and formamide acetate (3.8 g, 36.4 mmoles) in methanol (30 ml) is heated under reflux for 3 hours. The reaction mixture is cooled to room temperature and a saturated aqueous solution of sodium bicarbonate (30 ml) and water (30 ml) are added. The resulting precipitate is filtered and dried under vacuum to obtain the title compound.
  • Step G: Methyl 3-allyl-4-fluoro-5-(3-fluoro-4-iodophenylamino)-6-methoxycarbony-1-methyl-3H-benzoimidazol-1-ium iodide:
  • Figure imgb0058
  • A mixture of methyl 1-allyl-7-fluoro-6-(2-fluoro-4-iodophenylamino)-1H-benzo[d]imidazole-5-carboxylate (2.67 g, 5.7 mmoles) acetonitrile (10 ml) and iodomethane (5.5 ml, 83 mmoles) are stirred at 50 °C in a sealed tube for 12 hours. The reaction mixture is cooled and placed under reduced pressure to remove the excess of iodomethane. The crude material is poured into ether and the solid is isolated by filtration. The brown solid is dried under vacuum to obtain the title compound.
  • Step H: Methyl 4-fluoro-5-(3-fluoro-4-iodophenylamino)-1-methyl-1H-benzo[d]imidazole-6-carboxylate:
  • Figure imgb0059
  • A mixture of methyl 3-allyl-4-fluoro-5-(2-fluoro-4-iodophenylamino)-6-methoxycarbony-1-methyl-3H-benzoimidazol-1-ium iodide, triphenyl phosphine (0.62 g, 2.5 mmoles), palladium tetrakis (0.47 g, 0.48 mmoles) in dichloromethane (15 ml) is cooled at -10 °C. Pyrrolidine (0.65 ml) is added dropwise and the reaction mixture is stirred for 2 hours. Water is added and the mixture is extracted with dichloromethane. The organic phases are combined, washed with brine, dried Na2SO4 and concentrated under reduced pressure. The resulting solid is triturated with a mixture of Hexane/Ether (9/1) and isolated by filtration.
  • Step I: 4-Fluoro-5-(2-fluoro-4-iodophenylamino)-1-methyl-1H-benzo[d]imidazole-6-carboxylic acid:
  • Figure imgb0060
  • Methyl 4-fluoro-5-(2-fluoro-4-iodophenylamino)-1-methyl-1H-benzo[d]imidazole-6-carboxylate (2.15 g, 4.8 mmoles) is dissolved in a mixture of THF (30 ml), MeOH (10 ml) and H2O (5 ml) and LiOH (1.16 g, 4.8 mmoles) is added. The mixture is stirred at room temperature for 2 hours. Solvents are removed under reduced pressure and 1N HCl is added. The aqueous layer is extracted with chloroform. The organic layers are combined, washed with brine, dried (Na2SO4) and concentrated under reduced pressure to obtain the title compound.
  • Step J: 1-(2-fluoro-4-iodophenylamino)-8-fluoro-5-methyl-benzimidazo[6,5-d]imidazole:
  • Figure imgb0061
  • A mixture of 4-fluoro-5-(2-fluoro-4-iodophenylamino)-1-methyl-1H-benzo[d]imidazole-6-carboxylic acid (0.52 g, 1.2 mmoles), DPPA (0.52 ml, 2.4 mmoles) and triethyamine (1 ml, 06.0 mmoles) is refluxed for 3 hours. The solvent is removed under reduced pressure and the crude product is purified by flash chromatography using a gradient of EtOAc and hexane yielding the title compound.
  • Step K: 3-cyclopropanesulfonamide 1-(2-fluoro-4-iodophenylamino)-8-fluoro-5-methyl-benzimidazo[6,5-d]imidazole:
  • Figure imgb0062
  • The previous compound (Step J) (50 mg, 0.117 mmoles) is dissolved in THF (2 ml) and the mixture is cooled at -78 °C. LiHMDS (0.117 ml, 0.117 mmoles) is added dropwise followed by the addition of HMPA (0.5 ml). The mixture is warmed at room temperature and cyclopropyl sulfonyl chloride (0.013 ml, 0.14 mmoles) is added. After stirring at room temperature overnight, a saturated solution of ammonium chloride (5 ml) is added. The mixture is extracted with EtOAc (3 x 5 ml). The organic layers are combined, washed with brine, dried (Na2SO4) and concentrated under reduced pressure. The crude product is purified by flash chromatography using a gradient of EtOAc and hexane yielding to the title compound.
  • Step L: N-(4-fluoro-5-(3-fluoro-4-iodophenylamino)-1-methyl-1H-benzo[d]imidazol-6-yl)cyclopropanesulfonamide:
  • Figure imgb0063
  • The previous compound (Step K) (26 mg, 0.049 mmoles), potassium trimethylsilanolate (21 mg, 0.149 mmoles) dissolved in THF (2 ml) are stirred at room temperature overnight. Brine (3ml) is added and the mixture is extracted with EtOAc (3 x 5 ml). The organic layers are combined, dried (Na2SO4) and concentrated under reduced pressure. The crude product is purified by flash chromatography using a gradient of EtOAc and hexane yielding to the title compound.
  • Example 2: N-(4-fluoro-5-(2-fluoro-4-iodophenylamino)-1-methyl-1H-benzo[d]imidazol-6-yl)-1-(2-hydroxyethyl)cyclopropane-1-sulfonamide: Step A: 3-(1-bromocyclopropyl)ethanol:
  • Figure imgb0064
  • To a solution of neat diethyl zinc (3.3 ml, 3.977 g, 30 mmoles) in 100 ml anhydrous DCM was added very slowly trifluoroacetic acid (2.31 ml, 3.4188 g, 30 mmoles) dropwise at 0 °C. (Caution: Violent gas evolution, exothermic!). After completed addition of the TFA, the suspension was stirred for 20 min at the same temperature, followed by the addition of diiodomethane (2.45 ml, 8.134 g, 30.4 mmoles). It was further stirred at 0 °C for 20 min, and then a solution of 3-bromobut-3-en-1-ol (1 ml, 1.523 g, 10.1 mmoles) in 10 ml DCM was added at the same temperature. After complete addition, the mixture was warmed to room temperature and stirred for 4 hours. The mixture was quenched with 100 ml MeOH and 40 ml brine, and it was further stirred for 30 min. The solvents were reduced, and the residue extracted using CHCl3 / aq. NH4Cl. The organic layers were collected, washed with brine and water, and the solvent was removed to give 2-(1-bromocyclopropyl)-ethanol in sufficient purity (1.6564 g, 100%). 1H-NMR (500 MHz, CDCl3): δ = 3.90-3.83 (t, 2H), 1.91-1.87 (t, 2H), 1.71 (s, 1H, br), 1.14-1.09 (m, 2H), 0.83-0.79 (m, 2H).
  • Step B: (3-(1-bromocyclopropyl)ethoxy)(tert-butyl)dimethylsilane:
  • Figure imgb0065
  • To a solution of the cyclopropyl alcohol (Step A) (1.303 g, 7.95 mmoles) in 30 ml anhydrous DCM was added anhydrous pyridine (1.2 ml, 1,1736 g, 14.8 mmoles),TBSOTf (2.7 ml, 3.1077 g, 11.76 mol) and the solution was stirred at room temperature for 16 h. It was extracted with CHCl3 / brine and the organic fraction was dried with MgSO4. The solvent was reduced and the crude product purified using flash-column chromatography (CHCl3 / hexanes 1:10, Rf= 0.4). Yield: 0.796 g, 36%. 1H-NMR (500 MHz, CDCl3): δ= 3.95-3.75 (t, 2H), 1.95-1.85 (t, 2H), 1.15-1.05 (m, 2H), 0.95-0.80 (m, 11H), 0.15-0.05 (s, 6H).
  • Step C: 1-(2-(tert-butyldimethylsilyloxy)ethyl)cyclopropane-1-sulfonyl chloride:
  • Figure imgb0066
  • To a solution of the cyclopropyl bromide prepared in step B (1.1227 g, 4.04 mmoles) in 15 ml anhydrous diethyl ether was added a 1.7 M solution of t-BuLi in pentane (4.8 ml, 8.16 mmoles) at - 78 °C. The solution was stirred for 30 min at this temperature, and was then transferred via a transfer cannula into a solution of freshly distilled sulfuryl chloride (0.65 ml, 1.029 g, 8.1 mmoles) in 8 ml diethyl ether at - 78 °C. The yellow suspension was warmed to room temperature. The solvent was removed, and the residue was dried in vacuo to remove excessive sulfuryl chloride. Then, the residue was extracted two times with hexane, and after filtration the solvent was evaporated in vacuo to give the sulfonyl chloride in sufficient purity as a colorless oil. Yield: 870 mg (72%). 1H-NMR (300 MHz, CDCl3): δ=3.95-3.85 (t, 2H), 2.35-2.25 (t, 2H), 1.80-1.70 (m, 2H), 1.45-1.38 (m, 2H), 0.90 (s, 9H), 0.10 (s, 6H).
  • Step D: 1-(2-(tert-butyldimethylsilyloxy)ethyl)-1-(2-fluoro-4-iodophenylamino)-8-fluoro-5-methyl-benzimidazo[6,5-d]imidazolecyclopropane-3-sulfonamide:
  • Figure imgb0067
  • According to example 1 (step K), 1-(3-fluoro-4-iodophenylamino)-8-fluoro-5-methyl-benzimidazo[6,5-d]imidazole (81 mg, 0.19 mmoles) was reacted with the cyclopropylsulfonyl chloride (162 mg, 0.38 mmoles) prepared in step C to obtain the title product (46.6 mg, 36%). m/z = 687 [M-1]-.
  • Step E: 1-(2-(Tert-butyldimethylsilyloxy)ethyl)-N-(4-fluoro-5-(2-fluoro-4-iodophenylamino)-1-methyl-1H-benzo[d]imidazol-6-yl)cyclopropane-1-sulfonamide:
  • Figure imgb0068
  • According to example 1 (step L) the previous compound (46.6 mg, 0.067 mmoles) was reacted with potassium trimethylsilanolate (26 mg, 0.230 mmoles) in THF (1.5 ml) to obtain the title compound (39 mg, 87%).
  • Step F: N-(4-fluoro-5-(2-fluoro-4-iodophenylamino)-1-methyl-1H-benzo[d]imidazol-6-yl)-1-(2-hydroxyethyl)cyclopropane-1-sulfonamide:
  • Figure imgb0069
  • 1-(3-(Tert-butyldimethylsilyloxy)ethyl)-N-(4-fluoro-5-(3-fluoro-4-iodophenylamino)-1-methyl-1H-benzo[d]imidazol-6-yl)cyclopropane-1-sulfonamide (39 mg, 0.06 mmoles) was dissolved in THF (0.5 ml) and aqueous hydrochloric acid (1.2 N, 0.5 ml) was added at 0 °C. After stirring the reaction mixture for two hours, a saturated aqueous solution of sodium bicarbonate (5 ml) was added. The mixture was extracted with EtOAc (3 x 5 ml). The organic layers were combined, dried (Na2SO4) and concentrated under reduced pressure. The crude product was purified by flash chromatography using MeOH/CHCl3 (1/9) yielding to the title compound (27 mg, 83 %). m/z = 549 [M+1]-.
  • Example 3: N-(7-fluoro-6-(2-fluoro-4-iodophenylamino)-3-methylbenzo[d]isoxazol-5-yl)cyclopropanesulfonamide: Step A: 5-Bromo-3,4-difluoro-3-(3-fluorophenylamino)benzoic acid:
  • Figure imgb0070
  • To a solution of 2-fluoroaniline (4.27 ml, 44.3 mmol) in THF (30 ml) at -78 °C was added LiHMDS (66.6 ml, 1 M in THF, 66.6 mmol) dropwise. The reaction mixture was stirred for 10 min and a solution of 5-bromo-2,3,4-trifluorobenzoic acid (5.63g, 22.2 mmol) in THF (40 ml) was added to the mixture. The reaction was slowly warmed to room temperature and stirred at this temperature for 4 h. The mixture was concentrated, quenched with 10% HCl solution (70 ml), and extracted with EtOAc (2 x 200 ml). The combined organic solution was dried over MgSO4 and concentrated to dryness. Purification by trituration with boiling CH2Cl2 gave 4.40 g (57%) of 5-bromo-3,4-difluoro-2-(2-fluorophenylamino)benzoic acid as a yellow solid.
  • Step B: Methyl 5-bromo-3,4-difluoro-2-(2-fluorophenylamino)benzoate:
  • Figure imgb0071
  • To a solution of 5-bromo-3,4-difluoro-2-(2-fluorophenylamino)benzoic acid (3.0 g, 8.67 mmol) in a mixture of THF (15 ml) and MeOH (5 ml) was added (trimethylsilyl)diazomethane (5.63 ml, 2 M in Hexanes, 11.3 ml). After stirring for 4 h, the reaction was quenched with AcOH and diluted with EtOAc (100 ml). The organic solution was washed with H2O (50 ml) and brine (50 ml), dried over Na2SO4 and concentrated to dryness to give 2.83 g (91%) of methyl 5-bromo-3,4-difluoro-2-(2-fluorophenylamino)benzoate as a light yellow solid.
  • Step C: Methyl 3,4-difluoro-3-(3-fluorophenylamino)-5-((trimethylsilyl)ethynyl)benzoate:
  • Figure imgb0072
  • A mixture of methyl 5-bromo-3,4-difluoro-2-(2-fluorophenylamino)benzoate (2.70 g, 7.50 mmol), trimethylsilylacetylene (1.23 ml, 8.63 mmol), CuI (143 mg, 0.75 mmol), dichlorobis(triphenylphosphine)palladium (526 mg, 0.75 mmol) and i-Pr2NH (2.12 ml, 15.0 mmol) in THF (40 ml) was stirred at room temperature for 16 h. The reaction was concentrated and diluted with EtOAc (100 ml). The organic solution was washed with saturated aqueous NH4Cl (50 ml), dried over Na2SO4 and concentrated. Silica gel chromatography (EtOAc:Hexanes = 5:95) yielded 2.40 g (85%) of methyl 3,4-difluoro-2-(2-fluorophenylamino)-5-((trimethylsilyl)ethynyl)benzoate as a tan solid.
  • Step D: Methyl 5-acetyl-3,4-difluoro-2-(2-fluorophenylamino)benzoate:
  • Figure imgb0073
  • To a suspension of methyl 3,4-difluoro-2-(2-fluorophenylamino)-5-((trimethylsilyl)ethynyl)benzoate (1.20 g, 3.18 mmol) in 85% aqueous acetone (40 ml) were added HgSO4 (943 mg, 3.18 mmol) and H2SO4 (0.33 ml, 6.36 mmol). The reaction was heated at reflux for 24 h. The mixture was cooled to room temperature, concentrated, and diluted with EtOAc (100 ml). The organic solution was washed with H2O (50 ml), dried over Na2SO4 and concentrated. Silica gel chromatography (EtOAc:Hexanes = 10:90) yielded 832 mg (81%) of methyl 5-acetyl-3,4-difluoro-2-(2-fluorophenylamino)benzoate as a white solid.
  • Step E: Methyl 7-fluoro-6-(3-fluorophenylamino)-3-methylbenzo[d]isoxazole-5-carboxylate:
  • Figure imgb0074
  • To a solution of acetone oxime (3.17 g, 43.3 mmol) in THF (40 ml) was added t-BuOK (43.3 ml, 1 M in THF, 43.3 mmol). After stirring at room temperature for 30 min, the mixture was cooled to -78 °C. To the mixture was added a solution of methyl 5-acetyl-3,4-difluoro-3-(3-fluorophenylamino)benzoate (4.0 g, 12.4 mmol) in THF (70 ml) dropwise. After stirring at -78 °C for 15 min, the reaction was warmed to 0 °C and stirred at the temperature for 4 h. The reaction was quenched with saturated aqueous NH4Cl solution (200 ml) and extracted with EtOAc (2 x 200 ml). The organic solution was washed with brine (100 ml), dried over MgSO4 and concentrated. To the resulting oil was added a mixture of 5% HCl solution (100 ml) and MeOH (100 ml). The resulting suspension was heated at reflux for 1.5 h. The reaction was cooled to room temperature, concentrated to half volume, and diluted with EtOAc (200 ml). The organic solution was washed with brine (100 ml), dried over Na2SO4 and concentrated. Silica gel chromatography (EtOAc:Hexanes = 25:75) yielded 2.84 g (72%) of methyl 7-fluoro-6-(3-fluorophenylamino)-3-methylbenzo[d]isoxazole-5-carboxylate as a white solid.
  • Step F: Methyl 7-fluoro-6-(2-fluoro-4-iodophenylamino)-3-methylbenzo[d]isoxazole-5-carboxylate:
  • Figure imgb0075
  • To a suspension of methyl 7-fluoro-6-(2-fluorophenylamino)-3-methylbenzo[d]isoxazole-5-carboxylate (2.84 g, 8.93 mmol) in DMF (50 ml) were added N-iodosuccinimide (3.01 g, 13.4 mmol) and TFA (3 ml). After stirring at room temperature for 4 h, the reaction was quenched with saturated aqueous NH4Cl solution (100 ml) and extracted with EtOAc (2 x 100 ml). The combined organic solution was washed with brine (100 ml), dried over MgSO4 and concentrated. Silica gel chromatography (EtOAc:Hexanes = 25:75) yielded 3.77 g (95%) of methyl 7-fluoro-6-(3-fluoro-4-iodophenylamino)-3-methylbenzo[d]isoxazole-5-carboxylate as a tan solid.
  • Step G: 7-Fluoro-6-(2-fluoro-4-iodophenylamino)-3-methylbenzo[d]isoxazole-5-carboxylic acid:
  • Figure imgb0076
  • To a solution of methyl 7-fluoro-6-(3-fluoro-4-iodophenylamino)-3-methylbenzo[d]isoxazole-5-carboxylate (3.77 g, 8.49 mmol) in a mixture of THF (200 ml) and H2O (100 ml) was added a solution of LiOH (1.20 g, 50 mmol) in H2O (50 ml). After stirring at room temperature for 3 h, the reaction was concentrated, acidified with 1 M HCl solution, and extracted with EtOAc (2 x 100 ml). The organic solution was dried over Na2SO4 and concentrated to give 3.09 g (95%) of 7-fluoro-6-(2-fluoro-4-iodophenylamino)-3-methylbenzo[d]isoxazole-5-carboxylic acid as a light yellow solid.
  • Step H: 8-fluoro-1-(2-fluoro-4-iodophenyl)-5-methylbenzimidazolo[6,5-d][d]isoxazole:
  • Figure imgb0077
  • To a suspension of 7-fluoro-6-(2-fluoro-4-iodophenylamino)-3-methylbenzo[d]isoxazole-5-carboxylic acid (3.09 g, 8.08 mmol) in toluene (100 ml) was added triethylamine (3.37 ml, 24.2 mmol) and diphenylphosphoryl azide (2.18 ml, 10.1 mmol). The reaction was heated at reflux for 4 h. The mixture was cooled to room temperature and concentrated to dryness. Silica gel chromatography (EtOAc:Hexanes = 75:25) yielded 2.06 g (60%) of 8-fluoro-1-(2-fluoro-4-iodophenyl)-5-methylbenzimidazolo[6,5-d][d]isoxazole as a tan solid.
  • Step I: 3-(Cyclopropanesulfonyl)-8-fluoro-1-(2-fluoro-4-iodophenyl)-5-methylbenzimidazolo[6,5-d][d]isoxazole:
  • Figure imgb0078
  • To a solution of 8-fluoro-1-(2-fluoro-4-iodophenyl)-5-methylbenzimidazolo[6,5-d][d]isoxazole (200 mg, 0.47 mmol) in THF (15 ml) at -78 °C was added LiHMDS (0.52 ml, 1 M in THF, 0.52 mmol). The reaction mixture was warmed to 0 °C and stirred at the temperature for 10 min. To the reaction was added cyclopropylsulfonyl chloride (99 mg, 0.71 mmol) and the mixture was warmed to room temperature. After stirring at room temperature for 16 h, the reaction was quenched with saturated aqueous NH4Cl solution (20 ml) and extracted with EtOAc (2 x 30 ml). The combined organic solution was washed with brine (30 ml), dried over MgSO4 and concentrated. Silica gel chromatography (EtOAc:Hexanes = 25:75) yielded 222 mg (89%) of 3-(cyclopropanesulfonyl)-8-fluoro-1-(2-fluoro-4-iodophenyl)-5-methylbenzimidazolo[6,5-d][d]isoxazole as a white solid.
  • Step J: N-(7-fluoro-6-(3-fluoro-4-iodophenylamino)-3-methylbenzo[d]isoxazol-5-yl)cyclopropanesulfonamide:
  • Figure imgb0079
  • To a solution of 3-(cyclopropanesulfonyl)-8-fluoro-1-(2-fluoro-4-iodophenyl)-5-methylbenzimidazolo[6,5-d][d]isoxazole (139 mg, 0.26 mmol) in THF (15 ml) was added potassium trimethylsilanolate (101 mg, 0.78 mmol). The resulting cloudy solution was stirred at room temperature for 16 h. The reaction was quenched with saturated aqueous NH4Cl solution (20 ml) and extracted with EtOAc (2 x 30 ml). The combined organic solution was washed with brine (30 ml), dried over MgSO4 and concentrated. Silica gel chromatography (EtOAc:Hexanes = 50:50) yielded 121 mg (92%) of N-(7-fluoro-6-(3-fluoro-4-iodophenylamino)-3-methylbenzo[d]isoxazol-5-yl)cyclopropanesulfonamide as a white solid. 1H NMR (DMSO-d 6, 400 MHz): δ 0.74 (m, 2H), 0.81 (m, 2H), 2.58 (m, 4H), 6.60 (m, 1H), 7.32 (d, J = 8 Hz, 1H), 7.59 (d, J = 12 Hz, 1H), 7.68 (s, 1H), 7.80 (s, 1H), 9.48 (s, 1H).
  • Example 4: 1-(2-hydroxyethyl)-N-(7-fluoro-6-(2-fluoro-4-iodophenylamino)-3-methylbenzo[d]isoxazol-5-yl)cyclopropane-1-sulfonamide: Step A: 3-(3-(tert-butyldimethylsilyloxy)ethyl)cyclopropanesulfonyl)-8-fluoro-1-(3-fluoro-4-iodophenyl)-5-methylbenzimidazolo[6,5-d][d]isoxazole:
  • Figure imgb0080
  • To a solution of 8-fluoro-1-(2-fluoro-4-iodophenyl)-5-methylbenzimidazolo[6,5-d][d]isoxazole (example 3, step H) (150 mg, 0.35 mmol) in THF (15 ml) at -78 °C was added LiHMDS (0.39 ml, 1 M in THF, 0.39 mmol). The reaction mixture was stirred at -78 °C for 30 min and 1-(2-(tert-butyldimethylsilyloxy)ethyl)cyclopropane-1-sulfonyl chloride (144 mg, 0.48 mmol) was added to the mixture. The reaction was slowly warmed to room temperature and stirred at the temperature for 16 h. The reaction was quenched with saturated aqueous NH4Cl solution (20 ml) and extracted with EtOAc (2 x 30 ml).The combined organic solution was washed with brine (30 ml), dried over MgSO4 and concentrated. Silica gel chromatography (EtOAc:Hexanes = 10:90) yielded 43 mg (18%) of 3-(2-(tert-butyldimethylsilyloxy)ethyl)cyclopropanesulfonyl)-8-fluoro-1-(2-fluoro-4-iodophenyl)-5-methylbenzimidazolo[6,5-d][d]isoxazole as a light yellow oil.
  • Step B: 1-(3-(tert-butyldimethylsilyloxy)ethyl)-N-(7-fluoro-6-(3-fluoro-4-iodophenylamino)-3-methylbenzo[d]isoxazol-5-yl)cyclopropane-1-sulfonamide:
  • Figure imgb0081
  • To a solution of 3-(2-(tert-butyldimethylsilyloxy)ethyl)cyclopropanesulfonyl)-8-fluoro-1-(2-fluoro-4-iodophenyl)-5-methylbenzimidazolo[6,5-d][d]isoxazole (43 mg, 0.062 mmol) in THF (5 ml) was added potassium trimethylsilanolate (40 mg, 0.31 mmol). The resulting solution was stirred at room temperature for 24 h. The reaction was quenched with saturated aqueous NH4Cl solution (10 ml) and extracted with EtOAc (2 x 10 ml). The combined organic solution was washed with brine (10 ml), dried over MgSO4 and concentrated. Silica gel chromatography (EtOAc:Hexanes = 25:75) yielded 22 mg (53%) of 1-(2-(tert-butyldimethylsilyloxy)ethyl)-N-(7-fluoro-6-(2-fluoro-4-iodophenylamino)-3-methylbenzo[d]isoxazol-5-yl)cyclopropane-1-sulfonamide as a light yellow paste.
  • Step C: 1-(3-hydroxyethyl)-N-(7-fluoro-6-(2-fluoro-4-iodophenylamino)-3-methylbenzo[d]isoxazol-5-yl)cyclopropane-1-sulfonamide:
  • Figure imgb0082
  • To a solution of 1-(3-(tert-butyldimethylsilyloxy)ethyl)-N-(7-fluoro-6-(2-fluoro-4-iodophenylamino)-3-methylbenzo[d]isoxazol-5-yl)cyclopropane-1-sulfonamide (20 mg, 0.030 mmol) in THF (2 ml) was added HCl (0.10 ml, 1.2 N in H2O, 0.12 mmol). After stirring for 1 h, the reaction was quenched with saturated aqueous NaHCO3 solution (3 ml) and extracted with EtOAc (2 x 10 ml). The combined organic solution was dried over Na2SO4 and concentrated. Silica gel chromatography (EtOAc:Hexanes = 75:25) yielded 10 mg (61%) of 1-(3-hydroxyethyl)-N-(7-fluoro-6-(2-fluoro-4-iodophenylamino)-3-methylbenzo[d]isoxazol-5-yl)cyclopropane-1-sulfonamide as a white solid. 1H NMR (DMSO-d 6, 400 MHz): δ 0.75 (m, 2H), 0.87 (m, 2H), 1.84 (t, J = 8 Hz, 2H), 2.57 (s, 3H), 3.38 (m, 2H), 4.52 (s, 1H), 6.59 (m, 1H), 7.34 (d, J = 8 Hz, 1H), 7.60 (dd, J = 4, 8 Hz, 1H), 7.68 (s, 1H), 7.84 (s, 1H), 9.58 (s, 1H).
  • Example 5: 1-(2,3-dihydroxypropyl)-N-(7-fluoro-6-(2-fluoro-4-iodophenylamino)-3-methylbenzo[d]isoxazol-5-yl)cyclopropane-1-sulfonamide Step A: 3-(allylcyclopropanesulfonyl)-8-fluoro-1-(2-fluoro-4-iodophenyl)-5-methylbenzimidazolo[6,5-d][d]isoxazole:
  • Figure imgb0083
  • To a solution of 8-fluoro-1-(2-fluoro-4-iodophenyl)-5-methylbenzimidazolo[6,5-d][d]isoxazole (example 3, step H) (300 mg, 0.70 mmol) in THF (30 ml) at -78 °C was added LiHMDS (0.77 ml, 1 M in THF, 0.77 mmol). The reaction mixture was stirred at -78 °C for 30 min and 1-allylcyclopropane-1-sulfonyl chloride (253 mg, 1.40 mmol) was added to the mixture. The reaction was slowly warmed to room temperature and stirred at the temperature for 3 days. The reaction was quenched with saturated aqueous NH4Cl solution (40 ml) and extracted with EtOAc (2 x 50 ml). The combined organic solution was washed with brine (50 ml), dried over MgSO4 and concentrated. Silica gel chromatography (EtOAc:Hexanes = 25:75) yielded 109 mg (27%) of 3-(allylcyclopropanesulfonyl)-8-fluoro-1-(2-fluoro-4-iodophenyl)-5-methylbenzimidazolo[6,5-d][d]isoxazole as a white solid.
  • Step B: 1-allyl-N-(7-fluoro-6-(2-fluoro-4-iodophenylamino)-3-methylbenzo[d]isoxazol-5-yl)cyclopropane-1-sulfonamide:
  • Figure imgb0084
  • To a solution of 3-(allylcyclopropanesulfonyl)-8-fluoro-1-(2-fluoro-4-iodophenyl)-5-methylbenzimidazolo[6,5-d][d]isoxazole (133 mg, 0.23 mmol) in THF (10 ml) was added potassium trimethylsilanolate (36 mg, 0.28 mmol) at 0 °C. The resulting solution was stirred at 0 °C for 5 h. The reaction was quenched with saturated aqueous NH4Cl solution (10 ml) and extracted with EtOAc (2 x 10 ml). The combined organic solution was washed with brine (10 ml), dried over MgSO4 and concentrated. Silica gel chromatography (EtOAc:Hexanes = 50:50) yielded 53 mg (63%) of 1-allyl-N-(7-fluoro-6-(2-fluoro-4-iodophenylamino)-3-methylbenzo[d]isoxazol-5-yl)cyclopropane-1-sulfonamide as a light yellow paste.
  • Step C: 1-(2,3-dihydroxypropyl)-N-(7-fluoro-6-(2-fluoro-4-iodophenylamino)-3-methylbenzo[d]isoxazol-5-yl)cyclopropane-1-sulfonamide:
  • Figure imgb0085
  • To a solution of 1-allyl-N-(7-fluoro-6-(2-fluoro-4-iodophenylamino)-3-methylbenzo[d]isoxazol-5-yl)cyclopropane-1-sulfonamide (92 mg, 0.17 mmol) in THF (5 ml) were added N-methylmorporine N-oxide (20 mg, 0.17 mmol) and osmium tetraoxide (0.11 mL, 4 % in water, 0.017 mmol). After stirring for 16 h, the reaction was quenched with sodium sulfite solution (4 ml, 1M) and EtOAc (4 ml). The resulting mixture was stirred for 30 min and separated. The organic solution was dried over Na2SO4 and concentrated. Silica gel chromatography (MeOH:CH2Cl2 = 10:90) yielded 56 mg (57%) of 1-(2,3-dihydroxypropyl)-N-(7-fluoro-6-(2-fluoro-4-iodophenylamino)-3-methylbenzo[d]isoxazol-5-yl)cyclopropane-1-sulfonamide as a light purple paste. 1H NMR (DMSO-d 6, 400 MHz): δ 0.90 (m, 4H), 1.60 (m, 1H), 2.07 (m, 1H), 3.14 (m, 2H), 3.40 (m, 1H), 4.53 (t, 1H), 4.57 (d, 1H), 6.57 (m, 1H), 7.33 (d, J = 8 Hz, 1H), 7.59 (d, J = 11 Hz, 1H), 7.72 (s, 1H), 7.79 (s, 1H), 9.53 (s, 1H).
  • Example 6: N-(7-fluoro-6-(4-bromo-2-chlorophenylamino)-3-methylbenzo[d]isoxazol-5-yl)cyclopropanesulfonamide: Step A: 8-fluoro-1-(4-bromo-2-chlorophenyl)-5-methylbenzimidazolo[6,5-d][d]isoxazole:
  • Figure imgb0086
  • To a suspension of 7-fluoro-6-(4-bromo-2-chlorophenylamino)-3-methylbenzo[d]isoxazole-5-carboxylic acid (synthesized according to WO 2005/023759 ) (321 mg, 0.80 mmol) in toluene (20 ml) was added triethylamine (0.34 ml, 2.40 mmol) and diphenylphosphoryl azide (0.35 ml, 1.61 mmol). The reaction was heated at reflux for 4 h. The mixture was cooled to room temperature and concentrated to dryness. Silica gel chromatography (EtOAc:Hexanes = 75:25) yielded 259 mg (82%) of 8-fluoro-1-(4-bromo-2-chlorophenyl)-5-methylbenzimidazolo[6,5-d][d]isoxazole as a tan solid.
  • Step B: 3-(Cyclopropanesulfonyl)-8-fluoro-1-(4-bromo-2-chlorophenyl)-5-methylbenzimidazolo[6,5-d][d]isoxazole:
  • Figure imgb0087
  • To a solution of 8-fluoro-1-(4-bromo-2-chlorophenyl)-5-methylbenzimidazolo[6,5-d][d]isoxazole (39 mg, 0.098 mmol) in THF (4 ml) at -78 °C was added LiHMDS (0.15 ml, 1 M in THF, 0.15 mmol). The reaction mixture was stirred at -78 °C for 10 min and cyclopropylsulfonyl chloride (28 mg, 0.20 mmol) was added to the mixture. The reaction was slowly warmed to room temperature and stirred at the temperature for 3 h. The reaction was quenched with saturated aqueous NH4Cl solution (10 ml) and extracted with EtOAc (2 x 10 ml). The combined organic solution was washed with brine (10 ml), dried over MgSO4 and concentrated. Silica gel chromatography (EtOAc:Hexanes = 50:50) yielded 21 mg (43%) of 3-(cyclopropanesulfonyl)-8-fluoro-1-(4-bromo-2-chlorophenyl)-5-methylbenzimidazolo[6,5-d][d]isoxazole as a white solid.
  • Step C: N-(7-fluoro-6-(4-bromo-2-chlorophenylamino)-3-methylbenzo[d]isoxazol-5-yl)cyclopropanesulfonamide:
  • Figure imgb0088
  • To a solution of 3-(cyclopropanesulfonyl)-8-fluoro-1-(4-bromo-2-chlorophenyl)-5-methylbenzimidazolo[6,5-d][d]isoxazole (21 mg, 0.042 mmol) in THF (3 ml) was added potassium trimethylsilanolate (16 mg, 0.13 mmol). The reaction was stirred at room temperature for 16 h, quenched with saturated aqueous NH4Cl solution (10 ml) and extracted with EtOAc (2 x 10 ml). The combined organic solution was washed with brine (10 ml), dried over MgSO4 and concentrated. Silica gel chromatography (EtOAc:Hexanes = 75:25) yielded 20 mg (100%) of N-(7-fluoro-6-(4-bromo-2-chlorophenylamino)-3-methylbenzo[d]isoxazol-5-yl)cyclopropanesulfonamide as a white solid. 1H NMR (DMSO-d 6, 400 MHz): δ 0.83 (m, 4H), 2.60 (m, 4H), 6.68 (dd, J = 4, 8 Hz, 1H), 7.29 (dd, J = 4, 8 Hz, 1H), 7.59 (s, 1H), 7.68 (d, J = 4 Hz, 1H), 7.72 (s, 1H), 9.68 (s, 1H).
  • Example 7: N-(7-fluoro-6-(2-fluoro-4-iodophenylamino)-3-methylbenzo[c]isoxazol-5-yl)cyclopropanesulfonamide: Step A: Methyl 7-fluoro-6-(2-fluorophenylamino)-3-methylbenzo[c]isoxazole-5-carboxylate:
  • Figure imgb0089
  • To a solution of methyl 5-acetyl-3,4-difluoro-2-(2-fluorophenylamino)benzoate (example 3, step D) (1.50 g, 4.64 mmol) in a mixture of acetone (36 ml) and H2O (12 ml) was added NaN3 (452 mg, 6.96 mmol). The reaction was heated at 65 °C for 16 h and cooled to room temperature. Acetone was concentrated and the resulting mixture was extracted with EtOAc (2 x 50 ml). The combined organic solution was washed with brine (50 ml), dried over MgSO4 and concentrated to give a yellow solid. To the solid was added H2O (24 ml) and the reaction was heated at reflux for 3 h. The reaction was cooled to room temperature, filtered, and washed with H2O. The resulting solid was dried under high vacuum with P2O5 at 65°C for 24 h to give 1.31 g (89%) of methyl 7-fluoro-6-(2-fluorophenylamino)-3-methylbenzo[c]isoxazole-5-carboxylate as a yellow solid.
  • Step B: methyl 7-fluoro-6-(2-fluoro-4-iodophenylamino)-3-methylbenzo[c]isoxazole-5-carboxylate:
  • Figure imgb0090
  • To a suspension of methyl 7-fluoro-6-(2-fluorophenylamino)-3-methylbenzo[c]isoxazole-5-carboxylate (1.31 g, 4.12 mmol) in DMF (40 ml) was added N-iodosuccinimide (1.39 g, 6.17 mmol) and TFA (2.4 ml). After stirring at room temperature for 4 h, the reaction was quenched with saturated aqueous NH4Cl solution (100 ml) and extracted with EtOAc (2 x 100 ml). The combined organic solution was washed with brine (100 ml), dried over MgSO4 and concentrated. Silica gel chromatography (EtOAc:Hexanes = 25:75) yielded 1.71 g (93%) of methyl 7-fluoro-6-(3-fluoro-4-iodophenylamino)-3-methylbenzo[c]isoxazole-5-carboxylate as a brown solid.
  • Step C: 7-fluoro-6-(2-fluoro-4-iodophenylamino)-3-methylbenzo[c]isoxazole-5-carboxylic acid:
  • Figure imgb0091
  • To a solution of methyl 7-fluoro-6-(2-fluoro-4-iodophenylamino)-3-methylbenzo[c]isoxazole-5-carboxylate (214 mg, 0.48 mmol) in a mixture of THF (10 ml) and H2O (5 ml) was added a solution of LiOH (57.5 mg, 2.40 mmol) in H2O (2.4 ml). After stirring at room temperature for 4 h, the reaction was concentrated, acidified with 1 M HCl solution, and extracted with EtOAc (2 x 20 ml). The organic solution was dried over MgSO4 and concentrated to give 184 mg (100%) of 7-fluoro-6-(2-fluoro-4-iodophenylamino)-3-methylbenzo[c]isoxazole-5-carboxylic acid as an orange solid.
  • Step D: 8-fluoro-1-(2-fluoro-4-iodophenyl)-5-methylbenzimidazolo[6,5-d][c]isoxazole:
    Figure imgb0092
  • To a suspension of 7-fluoro-6-(2-fluoro-4-iodophenylamino)-3-methylbenzo[c]isoxazole-5-carboxylic acid (184 mg, 0.48 mmol) in toluene (10 ml) was added triethylamine (0.20 ml, 1.44 mmol) and diphenylphosphoryl azide (0.21 ml, 0.96 mmol). The reaction was heated at reflux for 4 h. The mixture was cooled to room temperature and concentrated to dryness. Silica gel chromatography (EtOAc:Hexanes = 75:25) yielded 132 mg (64%) of 8-fluoro-1-(2-fluoro-4-iodophenyl)-5-methylbenzimidazolo[6,5-d][c]isoxazole as a tan solid.
  • Step E: 3-(cyclopropanesulfonyl)-8-fluoro-1-(2-fluoro-4-iodophenyl)-5-methylbenzimidazolo[6,5-d][c]isoxazole:
  • Figure imgb0093
  • To a solution of 8-fluoro-1-(3-fluoro-4-iodophenyl)-5-methylbenzimidazolo[6,5-d][c]isoxazole (124 mg, 0.29 mmol) in THF (10 ml) at -78 °C was added LiHMDS (0.35 ml, 1 M in THF, 0.35 mmol). The reaction mixture was stirred at -78 °C for 10 min and cyclopropylsulfonyl chloride (61 mg, 0.44 mmol) was added to the mixture. The reaction is slowly warmed to room temperature and stirred at the temperature for 16 h. The reaction was quenched with saturated aqueous NH4Cl solution (20 ml) and extracted with EtOAc (2 x 20 ml). The combined organic solution was washed with brine (20 ml), dried over MgSO4 and concentrated. Silica gel chromatography (EtOAc:Hexanes = 25:75) yielded 122 mg (79%) of 3-(cyclopropanesulfonyl)-8-fluoro-1-(2-fluoro-4-iodophenyl)-5-methylbenzimidazolo[6,5-d][c]isoxazole as a yellow solid.
  • Step F: N-(7-fluoro-6-(3-fluoro-4-iodophenylamino)-3-methylbenzo[c]isoxazol-5-yl)cyclopropanesulfonamide:
  • Figure imgb0094
  • To a solution of 3-(cyclopropanesulfonyl)-8-fluoro-1-(2-fluoro-4-iodophenyl)-5-methylbenzimidazolo[6,5-d][c]isoxazole (120 mg, 0.23 mmol) in THF (15 ml) was added potassium trimethylsilanolate (87 mg, 0.68 mmol). The reaction was stirred at room temperature for 16 h, quenched with saturated aqueous NH4Cl solution (20 ml), and extracted with EtOAc (2 x 30 ml). The combined organic solution was washed with brine (30 ml), dried over MgSO4, and concentrated. Silica gel chromatography (EtOAc:Hexanes = 50:50) yielded 106 mg (92%) of N-(7-fluoro-6-(3-fluoro-4-iodophenylamino)-3-methylbenzo[c]isoxazol-5-yl)cyclopropanesulfonamide as a light yellow solid. 1H NMR (DMSO-d 6, 400 MHz): δ 0.73 (m, 2H), 0.85 (m, 2H), 2.63 (m, 1H), 2.85 (s, 3H), 6.65 (m, 1H), 7.36 (d, J= 8 Hz, 1H), 7.58 (s, 1H), 7.62 (m, 2H), 9.39 (s, 1H).
  • Example 8: N-(7-fluoro-6-(2-fluoro-4-iodophenylamino)-3-methylbenzo[c]isoxazol-5-yl)-1-(2-hydroxyethyl)cyclopropane-1-sulfonamide:
  • Figure imgb0095
  • This compound is synthesized from 8-fluoro-1-(2-fluoro-4-iodophenyl)-5-methylbenzimidazolo[6,5-d][c]isoxazole according to example 4 (step A, B and C).
  • Example 9: N-(7-(4-bromo-2-chlorophenylamino)-8-chloroimidazo[1,2-a]pyridin-6-yl)cyclopropanesulfonamide: Step A: Methyl 4,6-dichloronicotinate:
  • Figure imgb0096
  • To a suspension of methyl 4,6-dihydroxynicotinate (19.99 g, 118.3 mmol) in 100 ml POCl3 was added NEt3 (15.2 ml) dropwise at 0 °C over a period of 20 min. The thick suspension was warmed to room temperature and then to 80 °C for 3 h with stirring. Then, it was cooled down to room temperature and stirred for an additional 18 h. The mixture was poured into 1.51 crushed ice, extracted with EtOAc (3 x 150 ml), dried (Na2SO4) and concentrated under reduced pressure. The crude product was filtrated over silica using DCM to obtain the title compound as a yellow oil (19.2g, 79%). Rf (EtOAc/Hexanes 1: 1) = 0.55. 1H-NMR (500 MHz, CDCl3): δ = 8.82 (s, 1H), 7.45 (s, 1H), 3.90 (s, 3H).
  • Step B: 4,6-Dichloronicotinic acid:
  • Figure imgb0097
  • To a solution of methyl 4,6-dichloronicotinate in a mixture of THF (400 ml), MeOH (100 ml) and H2O (100 ml) was added a solution of NaOH (10 g) in 40 ml H2O. The mixture was stirred for 40 min. at room temp. Then, the solvents were reduced and it was acidified with conc. HCl to a pH of about 2. It was extracted using a mixture of Et2O/EtOAc and the organic layer was dried with Na2SO4. The solvents were removed and the residue dried in vacuo to obtain the title compound as a white solid (12.3 g, 69%). Rf (CHCl3/MeOH 10:1) = 0.85. 1H-NMR (300 MHz, DMSO-D6): δ = 8.80 (s, 1H), 7.90 (s, 1H).
  • Step C: 4-(4-Bromo-2-chlorophenylamino)-6-chloronicotinic acid:
  • Figure imgb0098
  • To a solution of 2-chloro-4-bromoaniline (6.776 g, 33.1 mmol) in 50 ml anhydrous THF was added LHMDS (50 ml, 50 mmol, 1M/THF) over a period of 15 min at -78 °C. It was stirred for an additional 30 min., then a solution of 4,6-dichloronicotinic acid (3 g, 15.7 mmol) in 50 ml THF was added dropwise. After complete addition the mixture was gradually allowed to warm to room temperature and the reaction mixture was stirred for an additional 12 hours. The mixture was quenched with H2O and 1N HCl was added (pH 0-1) followed by brine. It was extracted using THF and dried with Na2SO4. The solvent was removed and the solid crude product was suspended in 200 ml EtOAc. The suspension was heated with stirring at the reflux temperature for 5 min. It was cooled to room temperature and the precipitate was filtered and washed with EtOAc and dried at 50 °C for 5 h in oil pump vacuo to obtain the title compound as an off-white solid (4.05 g, 72%). Rf (CHCl3/MeOH 10:1) = 0.3. 1H-NMR (500 MHz, DMSO-D6): δ = 10.12 (s, 1H), 8.65 (s, 1H), 7.88 (s, 1H), 7.62-7.57 (dd, 1H), 7.55-7.48 (dd, 1H), 6.67 (s, 1H).
  • Step D: 4-(4-Bromo-3-chlorophenylamino)-5,6-dichloronicotinic acid:
  • Figure imgb0099
  • To a thick suspension of 4-(4-bromo-2-chlorophenylamino)-6-chloronicotinic acid (13.25 g, 36.8 mmol) in 350 ml DMF was added NCS (4.94 g, 37 mmol) and the suspension was stirred at room temperature for 3 days. Since the reaction did not go to completion (NMR control), an additional 1.987 g NCS was added and the mixture was stirred for 18 h, upon which the reaction went to completion. The mixture was poured into a solution of 20 g sodium metabisulfite in H2O (11) and the thick suspension was rest for 30 min. It was filtered, and the precipitate was washed with water, then with hexanes and it was dried in oil pump vacuo at 50 °C for 24 hours to obtain the desired compound as a white solid (13.11 g, 90%). 1H-NMR (500 MHz, DMSO-D6): δ = 9.90 (s, br, 1H), 8.63 (s, 1H), 7.74 (s, 1H), 7.43-7.42 (dd, 1H), 6.99-6.97 (dd, 1H).
  • Step E: Methyl 4-(4-bromo-2-chlorophenylamino)-5,6-dichloronicotinate:
  • Figure imgb0100
  • To a suspension of 4-(4-bromo-2-chlorophenylamino)-5,6-dichloronicotinic acid (4.3975 g, 11.2 mmol) in a mixture 70 ml MeOH and 70 ml toluene was added TMS-diazomethane (10 ml, 20 mmol, 2M/hexanes) in small portions at room temperature. At the end of the addition the suspension turned into a solution, and then a precipitate was formed again. The suspension was diluted with hexanes and filtered. And the precipitate was washed with hexanes and dried in vacuo to obtain the title compound 3.03 g (66%).
  • Rf (EtOAc/Hexanes 1:3) = 0.5. 1H-NMR (500 MHz, DMSO-D6): δ = 9.29 (s, 1H), 8.55 (s, 1H), 7.80-7.79 (d, 1H), 7.45-7.40 (dd, 1H), 7.05-7.03 (d, 1H), 3.65 (s, 3H).
  • Step F: Methyl 6-azido-4-(4-bromo-3-chlorophenylamino)-5-chloronicotinate:
  • Figure imgb0101
  • Methyl 4-(4-bromo-2-chlorophenylamino)-5,6-dichloronicotinate (1.690 g, 4.14 mmol) was suspended in 30 ml DMF and the suspension was heated until everything went into solution. It was cooled down to room temperature and to that solution was added solid sodium azide (0.533 g, 8.2 mmol). The yellow solution was stirred for 17 h at room temperature and was diluted with water. The precipitate was filtered, washed with water and hexanes and dried in oil-pump vacuo at 50 °C for 2 h to obtain the title compound as a light-brown solid (1.5422 g, 90%). 1H-NMR (300 MHz, DMSO-D6): δ = 9.85 (s, 1H), 8.90 (s, 1H), 7.80-7.79 (d, 1H), 7.46-7.40 (dd, 1H), 6.95-6.90 (d, 1H), 3.80 (s, 3H).
  • Step G: Methyl 6-amino-4-(4-bromo-2-chlorophenylamino)-5-chloronicotinate:
  • Figure imgb0102
  • To a suspension of methyl 6-azido-4-(4-bromo-2-chlorophenylamino)-5-chloronicotinate (316 mg, 0.76 mmol) in a mixture of DCM (6 ml) and AcOH (2 ml) was added Zn dust (255 mg, 3.9 mmol) in small portions. After complete addition it was furthers stirred at room temperature for 30 min. Then, the mixture was poured into EtOAc (30 ml), washed with H2O, saturated sodium bicarbonate solution and brine. The organic fractions were dried with Na2SO4 and the solvents were removed and the product was dried in oil-pump vacuo (246 mg, 83%). Rf (CHCl3/MeOH 30:1) = 0.3. 1H-NMR (500 MHz, DMSO-D6): δ=9.17 (s, 1H), 8.45 (s, 1H), 7.70-7.70 (d, 1H), 7.38-7.36 (dd, 1H), 7.1 (s, 1H, br), 6.68-6.67 (d, 1H), 3.76 (s, 3H).
  • Step H: Methyl 7-(4-bromo-2-chlorophenylamino)-8-chloroimidazo[1,2-a]pyridine-6-carboxylate:
  • Figure imgb0103
  • To a suspension of methyl 6-amino-4-(4-bromo-2-chlorophenylamino)-5-chloronicotinate (966 mg, 2.48 mmol) in 13 ml DMF was added chloroacetaldehyde (50% aq. solution, 0.32 ml, 396 mg, 2.52 mmol) and the mixture was heated in a closed vessel to 80 °C for 3 h. The dark solution was cooled to room temperature and the DMF was removed by vacuum distillation. The residue was dissolved in THF and directly purified by column chromatography (EtOAc/hexanes 10:1 + 2% TEA) which delivered the desired product (638 mg, 62 %). Rf (EtOAc/Hexanes 10:1) = 0.4. 1H-NMR (500 MHz, MeOH-D3): δ = 9.16 (s, 1H), 7.86-7.85 (d, 1H), 7.54-7.53 (d, 1H), 7.50-7.48 (d, 1H), 7.19-7.17 (dd, 1H), 6.51-6.50 (d, 1H), 3.82 (s, 3H).
  • Step I: 7-(4-Bromo-2-chlorophenylamino)-8-chloroimidazo[1,2-a]pyridine-6-carboxylic acid:
  • Figure imgb0104
  • To a solution of methyl 7-(4-bromo-2-chlorophenylamino)-8-chloroimidazo[1,2-a]pyridine-6-carboxylate (366 mg, 0.81 mmol) in a mixture of MeOH (30 ml) and THF (10 ml) was added 10 ml of 1M aq. NaOH solution and the solution was stirred for 18 hours at room temperature. The solvents were reduced in vacuo, and it was extracted using 1M HCl / EtOAc / THF. The organic layers were washed with brine and dried with Na2SO4, and the solvents were removed in vacuo to give the crude product which was used without further purification (200 mg, 62%). 1H-NMR (500 MHz, DMSO-D6): δ = 9.33 (s, 1H), 8.11 (s, 1H), 7.69-7.68 (d, 1H), 7.35-7.33 (dd, 1H), 6.67-6.65 (d, br, 1H).
  • Step J: 3-(4-Bromo-3-chlorophenyl)-4-chloro-1H-diimidazo[1,3-a:4',5'-d]pyridin-2(3H)-one:
  • Figure imgb0105
  • To a suspension of 7-(4-bromo-2-chlorophenylamino)-8-chloroimidazo[1,2-a]pyridine-6-carboxylic acid (152mg, 0.35 mmol) (HCl salt) in 5 ml EtOAc was added NEt3 (0.14 ml, 102 mg, 1 mmol) and the mixture was stirred for 5 min upon the suspension turned into a solution. The solvents were removed in vacuo and the residue was dried in oil-pump vacuo. This residue was suspended in 4.5 ml anhydrous toluene and it was ultrasonicated for 1 min. DPPA (0.1 ml, 124 mg, 0.44 mmol) was added and the suspension was heated with stirring under argon at 125 °C oil bath temperature to reflux for 5 h. The initial suspension turns into a dark solution after about 30 min. heating, and then a precipitate is formed after about another 30 min. reaction time. After 5 h the reaction mixture was cooled to room temperature and the volatiles were removed in vacuo. To the dark residue was washed with Et2O (ultrasonication) and again dried in oil pump vacuo. The crude product was used without purification for the next step in the same reaction vessel. Rf (CHCl3/MeOH 4:1) = 0.5
  • Step K: 3-(4-bromo-3-chlorophenyl)-4-chloro-1-(cyclopropylsulfonyl)-1H-diimidazo[1,2-a:4',5'-d]pyridin-2(3H)-one:
  • Figure imgb0106
  • To a solution of the crude product (Step J) in 3 ml anhydrous THF was added LHMDS (0.4 ml, 0.4 mmol, 1M/THF) at 0 °C and the mixture was stirred for 5 min.. The cyclopropyl sulfonamide (83 mg, 0.59 mol) was added neat and the mixture was warmed to room temperature and stirred for 15 h. The mixture was quenched with brine, diluted with H2O and extracted using a mixture of EtOAc/THF (3:1). The organic phase was washed with brine and dried with Na2SO4. Subsequent column chromatography (100% EtOAc) gave the title product as a brown oil (36 mg, 20% over the 2 last steps). Rf (100% EtOAc) = 0.35. 1H-NMR (500 MHz, MeOH-D3): δ = 8.80 (s, 1H), 7.89 (s, 1H), 7.86-7.85 (d, 1H), 7.68-7.66 (dd, 1H), 7.60-7.58 (d, 1H), 7.46 (s, 1H), 1.51-1.37 (m, 2H), 1.23-1.15 (m, 2H).
  • Step L: N-(7-(4-bromo-3-chlorophenylamino)-8-chloroimidazo[1,2-a]pyridin-6-yl)cyclopropanesulfonamide:
  • Figure imgb0107
  • To a solution of N-(7-(4-bromo-2-chlorophenylamino)-8-chloroimidazo[1,2-a]pyridin-6-yl)cyclopropanesulfonamide (25 mg, 0.05 mmol) was added solid KOTMS (21 mg, 0.16 mmol) and the cloudy solution was stirred at 40 °C for 2.5 h. It was quenched with aqueous NH4Cl, extracted with EtOAc and dried with Na2SO4. The crude product was purified by column chromatography (CHCl3/MeOH 20:1) to give the title compound as a brown paste (11 mg, 47%). Rf (CHCl3/MeOH 15:1) = 0.25. 1H-NMR (500 MHz, CDCl3): δ = 8.46 (s, 1H), 7.70 (s, 1H), 7.66 (s, 1H), 7.56-7.55 (d, 1H), 7.18-7.16 (dd, 1H), 6.30-6.29 (d, 1H), 6.45 (s, br, 1H), 6.10 (s, br, 1H), 2.43-2.39 (m, 1H), 1.13-1.10 (m, 2H), 1.00-0.93 (m, 2H).
  • Example 10: N-(7-(4-bromo-2-chlorophenylamino)-8-chloroimidazo[1,2-a]pyridin-6-yl)-1-(2-hydroxyethyl)cyclopropane-1-sulfonamide:
  • Figure imgb0108
  • This compound was synthesized from 3-(4-bromo-2-chlorophenyl)-4-chloro-1-(cyclopropylsulfonyl)-1H-diimidazo[1,2-a:4',5'-d]pyridin-2(3H)-one (see example 9) according to example 4 (step A, B and C). 1H NMR (400 MHz, MeOD) δ ppm 0.87 - 0.98 (m, 2 H) 1.11 - 1.24 (m, 2 H) 1.99 - 2.13 (m, 2 H) 3.35 (s, 2 H) 3.71 (s, 3 H) 6.46 (d, J=8.71 Hz, 1 H) 7.24 (d, J=2.28 Hz, 1 H) 7.54 - 7.67 (m, 1 H) 7.95 (d, J=1.45 Hz, 1 H) 8.70 (s, 1 H).m/z = 519 [M-1]-.
  • Example 11: N-(8-chloro-7-(2-fluoro-4-iodophenylamino)imidazo[1,2-a]pyridin-6-yl)cyclopropanesulfonamide:
  • Figure imgb0109
  • This compound is synthesized from methyl 6-amino-5-chloro-4-(2-fluoro-4-iodophenylamino]nicotinate: (see below) according to example 9 (step H through L).
  • Methyl 6-amino-5-chloro-4-(2-fluoro-4-iodophenylamino)nicotinate: Step A: 6-Dichloro-4-(2-fluoro-4-iodophenylamino)nicotinic acid:
  • Figure imgb0110
  • To a solution of 2-fluoro-4-iodoaniline (11.3 g, 50.3 mmol) in 85 ml anhydrous THF is added LiHMDS (83 ml, 83 mmol, 1M/THF) over a period of 30 min at -78°C. It is stirred for another 30 min, then a solution of methyl 4,6-dichloronicotinate (step B, example 9) (5.00 g, 26.2 mmol) in 85 ml THF is added dropwise. After complete addition the mixture is gradually allowed to warm to room temperature and stirred for another 18 hrs. It is quenched with H2O, then 1N HCl is added to (pH = 1) followed by brine. It is extracted using THF and dried with Na2SO4. The solvents are removed and the crude solid is suspended in 300 ml of EtOAc. The suspension is heated with stirring at the reflux temperature for 5 min. It is cooled to room temperature and the precipitate is filtered and washed with EtOAc and dried at 50°C for 5 h in oil pump vacuo.
  • Step B: 5,6-Dichloro-4-(2-fluoro-4-iodophenylamino)nicotinic acid:
  • Figure imgb0111
  • This compound is synthesized from 6-dichloro-4-(2-fluoro-4-iodophenylamino)nicotinic acid (8.1 g, 23.5 mmol) according to example 9, step D. The title compound is obtained.
  • Step C: Methyl 5,6-dichloro-4-(2-fluoro-4-iodophenylamino)nicotinate:
  • Figure imgb0112
  • This compound is synthesized from 5,6-dichloro-4-(2-fluoro-4-iodophenylamino)nicotinic acid (3.00 g, 7.64 mmol) according to example 9, step E.
  • Step D: Methyl 6-azido-5-chloro-4-(2-fluoro-4-iodophenylamino)nicotinate:
  • Figure imgb0113
  • This compound is synthesized from methyl 5,6-dichloro-4-(2-fluoro-4-iodophenylamino)nicotinate (2.9 g, 6.5 mmol) according to example 9, step F.
  • Step E: Methyl 6-amino-5-chloro-4-(2-fluoro-4-iodophenylamino)nicotinate:
  • Figure imgb0114
  • This compound is synthesized from methyl 6-azido-5-chloro-4-(2-fluoro-4-iodophenylamino)nicotinate (2.65 g, 5.9 mmol) according to example 9, step G.
  • Example 12: N-(8-chloro-7-(2-fluoro-4-iodophenylamino)imidazo[1,2-a]pyridin-6-yl)-1-(2-hydroxyethyl)cyclopropane-1-sulfonamide:
  • Figure imgb0115
  • This compound was synthesized according to example 4 (step A, B and C) starting from 4-chloro-3-(2-fluoro-4-iodophenyl)-1H-diimidazo[1,2-a:4',5'-d]pyridin-2(3H)-one (Example 10). m/z = 549 [M-1]-. 1H NMR (400 MHz, MeOD) δ ppm 0.85 - 0.96 (m, 2 H) 1.12 - 1.24 (m, 2 H) 2.02 (t, J=6.63 Hz, 2 H) 3.69 (t, J=6.63 Hz, 2 H) 6.43 (t, J=8.71 Hz, 1 H) 7.32 (d, J=8.50 Hz, 1 H) 7.47 (dd, J=10.68, 1.76 Hz, 1 H) 7.60 (s, 1 H) 7.93 (s, 1 H) 8.65 (s, 1 H).
  • Example 13: N-(8-fluoro-7-(2-fluoro-4-iodophenylamino)imidazo[1,2-a]pyridin-6-yl)cyclopropanesulfonamide:
  • Figure imgb0116
  • This compound is synthesized according to example 9 (step H through L) starting from methyl 6-amino-5-fluoro-4-(2-fluoro-4-iodophenylamino)nicotinate (see below).
  • Methyl 6-amino-5-fluoro-4-(2-fluoro-4-iodophenylamino)nicotinate:
  • Figure imgb0117
  • To a solution of methyl 6-amino-5-chloro-4-(2-fluoro-4-iodophenylamino)nicotinate (2.0 g, 4.74 mmol) (Example 11, step E) in a mixture of MeOH/water (1/1) (20 ml) is added selectfluor (1.6 g, 4.74 mmol). The mixture is stirred at room temperature overnight, diluted with EtOAc and washed with 0.5 N HCl and brine. The organic extracts are dried (Na2SO4) and concentrated under reduced pressure. The crude material was purified by HPLC (reverse phase) to obtain the title compound.
  • Example 14: N-(7-(2-fluoro-4-iodophenylamino)-8-methylimidazo[1,2-a]pyridin-6-yl)cyclopropanesulfonamide:
  • Figure imgb0118
  • This compound was synthesized according to example 9 (step C through L) starting from methyl 4,6-dichloro-5-methylnicotinate (Journal of Heterocyclic Chemistry (1999), 36(4), 953-957). m/z = 485 [M-1]-. 1H NMR (DMSO-d 6, 400 MHz): δ 0.69 (m, 2H), 0.85 (m, 2H), 2.23 (s, 3H), 2.48 (m, 1H), 6.22 (m, 1H), 6.53 (m, 1H), 7.22 (d,1H), 7.45 (m, 1H), 7.55 (m, 2H), 8.00 (s, 1H), 8.56 (s, 1H).
  • Example 15: N-(7-(2-fluoro-4-iodophenylamino)-8-methylimidazo[1,2-a]pyridin-6-yl)-1-(2-hydroxyethyl)cyclopropane-1-sulfonamide
  • Figure imgb0119
  • This compound is synthesized according to example 4 (step A, B and C) starting from 3-(2-fluoro-4-iodophenyl)-4-methyl-1H-diimidazo[1,2-a:4',5'-d]pyridin-2(3H)-one.
  • Example 16: 1-(2,3-dihydroxypropyl)-N-(7-(2-fluoro-4-iodophenylamino)-8-methylimidazo[1,2-a]pyridin-6-yl)cyclopropane-1-sulfonamide
  • Figure imgb0120
  • This compound was synthesized according to example 5 (step A, B and C) starting from 3-(2-fluoro-4-iodophenyl)-4-methyl-1H-diimidazo[1,2-a:4',5'-d]pyridin-2(3H)-one. m/z = 559 [M-1]-. 1H NMR (DMSO-d 6, 400 MHz): δ 0.97 (m, 4H), 1.60 (m, 1H), 2.08 (m, 1H), 2.24 (s, 3H), 3.28 (m, 2H), 3.54 (m, 1H), 4.53 (m, 2H), 6.16 (m, 1H), 7.24 (m, 1H), 7.38 (s,1H), 7.54 (s, 2H), 7.97 (s, 1H), 8.57 (s, 1H), 9.34 (s, 1H).
  • Example 17: N-(7-(4-bromo-2-chlorophenylamino)-8-methylimidazo[1,2-a]pyridin-6-yl)cyclopropanesulfonamide
  • Figure imgb0121
  • This compound was synthesized according to example 9 (step C through L) starting from methyl 4,6-dichloro-5-methylnicotinate (Journal of Heterocyclic Chemistry (1999), 36(4), 953-957) m/z = 454 [M-1]-. 1H NMR (DMSO-d 6, 400 MHz): δ 0.78 (m, 2H), 0.86 (m, 2H), 2.22 (s, 3H), 6.29 (d, J = 8 Hz, 1H), 7.22 (d, J = 8 Hz, 1H), 7.31 (s, 1H), 7.56 (s, 1H), 7.64 (s, 1H), 8.01 (s, 1H), 8.60 (s, 1H), 9.34 (s, 1H).
  • Example 18: N-(7-(4-bromo-2-chlorophenylamino)-8-methylimidazo[1,2-a]pyridin-6-yl)-1-(2-hydroxyethyl)cyclopropane-1-sulfonamide
  • Figure imgb0122
  • This compound was synthesized according to example 4 (step A, B and C) starting from 3-(4-bromo-2-chlorophenyl)-4-methyl-1H-diimidazo[1,2-a:4',5'-d]pyridin-3(3H)-one. m/z = 499 [M-1]-. 1H NMR (DMSO-d 6,400 MHz): δ 0.88 (m, 4H), 1.94 (m, 2H), 2.24 (s, 3H), 3.48 (m, 2H), 4.54 (m, 1H), 6.30 (d, J = 8 Hz, 1H), 7.29 (d, J = 8 Hz, 1H), 7.37 (s, 1H), 7.59 (s, 1H), 8.03 (s, 1H), 8.61 (s, 1H), 9.35 (s, 1H).
  • Example 19 (not claimed): N-(8-chloro-7-(2-fluoro-4-iodophenylamino)-3-methyl-[1,2,4]diazole[4,3-a]pyridin-6-yl)cyclopropanesulfonamide: Step A: 5-chloro-4-(2-fluoro-4-iodophenylamino)hydrazinylnicotinic acid:
  • Figure imgb0123
  • Hydrazine monohydrate (0.2 ml, 4.12 mmol) is added to a solution of methyl 6-amino-5-chloro-4-(2-fluoro-4-iodophenylamino)nicotinate (example 10, step A, B and C) (2 g, 4.5 mmol) in N,N-dimethylacetamide (2.00 ml). After stirring at 90°C for 1h, the reaction mixture is cooled to room temperature and diluted with EtOAc. The organic layer is washed with H2O, brine, dried with Na2SO4 and concentrated under reduced pressure. The crude material is washed with DCM to obtain the title material.
  • Step B: 8-Chloro-7-(2-fluoro-4-iodophenylamino)-3-methyl-[1,2,4]triazolo[4,3-a]pyridine-6-carboxylic acid:
  • Figure imgb0124
  • Acetic anhydride (0.95 mmol) is added to a solution of 5-chloro-4-(2-fluoro-4-iodophenylamino)-6-hydrazinylnicotinic acid (0.4 g, 0.95 mmol) and triethylamine (1.9 mmol) in DCM (10 ml) at 0 °C. The reaction mixture is warmed to room temperature, stirred for an additional 10 min and POCl3 (0.95 mmol) is added. After stirring overnight at room temperature the mixture is refluxed for 3 days. It was diluted with EtOAc and NaHCO3 and stirred for 20 min. The organic layer is washed with H2O, brine, dried with Na2SO4 and concentrated under reduced pressure. The crude product is purified by column chromatography.
  • Step C: 9-chloro-8-(2-fluoro-4-iodophenyl)-3-methyl-6H-imidazo][4,5-d][1,2,4]triazolo[4,3-a]pyridin-7(8H)-one:
  • Figure imgb0125
  • Synthesize according to example 3, step H from 9-chloro-8-(2-fluoro-4-iodophenyl)-3-methyl-6H-imidazo[4,5-d][1,2,4]triazolo[4,3-a]pyridin-7(8H)-one (0.34, 0.76 mmol) to afford the title compound.
  • Step D: 9-chloro-6-(cyclopropylsulfonyl)-8-(2-fluoro-4-iodophenyl)-3-methyl-6H-imidazo[4,5-d][1,2,4]triazolo[4,3-a]pyridin-7(8H)-one:
  • Figure imgb0126
  • Synthesize according to example 3, step I from 9-chloro-8-(2-fluoro-4-iodophenyl)-3-methyl-6H-imidazo[4,5-d][1,2,4]triazolo[4,3-a]pyridin-7(8H)-one (100 mg, 0.22 mmol) to afford the title compound.
  • Step E: N-(8-chloro-7-(2-fluoro-4-iodophenylamino)-3-methyl-[1,2,4]triazolo[4,3-a]pyridin-6-yl)cyclopropanesulfonamide:
  • Figure imgb0127
  • Synthesize according to example 3, step J from 9-chloro-6-(cyclopropylsulfonyl)-8-(2-fluoro-4-iodophenyl)-3-methyl-6H-imidazo[4,5-d][1,2,4]triazolo[4,3-a]pyridin-7(8H)-one (98 mg, 018 mmol) to afford the title compound.
  • Example 20 (not claimed): N-(8-chloro-7-(2-fluoro-4-iodophenylamino)-3-methyl-[1,2,4]triazolo[4,3-a]pyridin-6-yl)-1-(2-hydroxyethyl)cyclopropane-1-sulfonamide:
  • Figure imgb0128
  • Synthesize according to example 4, step A, B and C from 9-chloro-8-(2-fluoro-4-iodophenyl)-3-methyl-6H-imidazo[4,5-d][1,2,4]triazolo[4,3-a]pyridin-7(8H)-one to afford the title compound.
  • Example 21 (not claimed): N-(7-chloro-6-(2-fluoro-4-iodophenylamino)-3-methyl-3a,7a-dihydro-3H-imidazo[4,5-b]pyridin-5-yl)cyclopropanesulfonamide:
  • Step A: 7-Chloro-6-(2-fluoro-4-iodophenylamino)-3-methyl-3H-imidazo[4,5-b]pyridine-5-carboxylic acid:
    Figure imgb0129
  • To a solution of 3-fluoro-4-iodo-aniline (2.9 g, 22.1 mmol) in THF (20 ml) at -78 °C is added LiHMDS (33.3 ml, 1 M in THF, 33.3 mmol) dropwise. The reaction mixture is stirred for 10 min and a solution of 6-bromo-7-chloro-3-methyl-3H-imidazo[4,5-b]pyridine-5-carboxylic acid ( WO 2005/051906 ) (5.63g, 10.1 mmol) in THF (30 ml) is added to the mixture. The reaction is slowly warmed to room temperature and stirred at this temperature for 16 h. The mixture is concentrated, quenched with 10% HCl solution (70 ml), and extracted with EtOAc (2 x 200 ml). The combined organic solution is dried over MgSO4 and concentrated to dryness. Purification by trituration with boiling CH2Cl2 gives the title compound.
  • Step B: 8-Chloro-1-(3-fluoro-4-iodophenyl)-5-methyl-3,5-dihydrodiimidazo[4,5-b:4',5'-e]pyridin-3(1H)-one:
  • Figure imgb0130
  • To a suspension of 7-chloro-6-(2-fluoro-4-iodophenylamino)-3-methyl-3H-imidazo[4,5-b]pyridine-5-carboxylic acid (2.75 g, 6.16 mmol) in toluene (80 ml) is added triethylamine (2.57 ml, 18.5 mmol) and diphenylphosphoryl azide (1.56 ml, 7.7 mmol). The reaction is heated at reflux for 4 h. The mixture is cooled to room temperature and concentrated to dryness. Silica gel chromatography yields the title compound.
  • Step C: 8-Chloro-3-(cyclopropylsulfonyl)-1-(2-fluoro-4-iodophenyl)-5-methyl-3,4a,5,7a-tetrahydrodiimidazo[4,5-b:4',5'-e]pyridin-2(1H)-one:
  • Figure imgb0131
  • To a solution of 8-chloro-1-(2-fluoro-4-iodophenyl]-5-methyl-3,5-dihydrodiimidazo[4,5-b:4',5'-e]pyridin-2(1H)-one (200 mg, 0.36 mmol) in THF (6 ml) at -78 °C is added LiHMDS (0.54 ml, 1 M in THF, 0.54 mmol). The reaction mixture is stirred at -78 °C for 10 min and cyclopropylsulfonyl chloride (100 mg, 0.72 mmol) is added to the mixture. The reaction is slowly warmed to room temperature and stirred at the temperature for 3 h. The reaction is quenched with saturated aqueous NH4Cl solution (10 ml) and extracted with EtOAc (2 x 10 ml). The combined organic solution is washed with brine (10 ml), dried over MgSO4 and concentrated. Silica gel chromatography yields the title compound.
  • Step D: N-(7-Chloro-6-(2-fluoro-4-iodophenylamino)-3-methyl-3a,7a-dihydro-3H-imidazo[4,5-b]pyridin-5-yl)cyclopropanesulfonamide:
  • Figure imgb0132
  • To a solution of 8-chloro-3-(cyclopropylsulfonyl)-1-(2-fluoro-4-iodophenyl)-5-methyl-3,4a,5,7a-tetrahydrodiimidazo[4,5-b:4',5'-e]pyridin-2(1H)-one (79 mg, 0.144 mmol) in THF (5 ml) is added potassium trimethylsilanolate (55 mg, 0.43 mmol). The reaction is stirred at room temperature for 16 h, quenched with saturated aqueous NH4Cl solution (10 ml) and extracted with EtOAc (2 x 10 ml). The combined organic solution is washed with brine (10 ml), dried over MgSO4 and concentrated. Silica gel chromatography yields the title compound.
  • Example 22 (not claimed): N-(7-chloro-6-(2-fluoro-4-iodophenylamino)-3-methyl-3a,7a-dihydro-3H-imidazo[4,5-b]pyridin-5-yl)-1-(2-hydroxyethyl)cyclopropane-1-sulfonamide: Step A: 3-(1-(2-(Tert-butyldimethylsilyloxy)ethyl)cyclopropylsulfonyl)-8-chloro-1-(2-fluoro-4-iodophenyl)-1-5-methyl-3,4a,5,7a-tetrahydrodiimidazo[4,5-b:4',5'-e]pyridin-2(1H)-one:
  • Figure imgb0133
  • To a solution of 8-chloro-1-(2-fluoro-4-iodophenyl)-5-methyl-3,5-dihydrodiimidazo[4,5-b:4',5'-e]pyridin-2(1H)-one (example 16, step B) (150 mg, 0.35 mmol) in THF (5 ml) at -78 °C is added LiHMDS (0.39 ml, 1 M in THF, 0.39 mmol). The reaction mixture is stirred at -78 °C for 30 min and 1-(2-(tert-butyldimethylsilyloxy)ethyl)cyclopropane-1-sulfonyl chloride (144 mg, 0.48 mmol) is added to the mixture. The reaction is slowly warmed to room temperature and stirred at the temperature for 16 h. The reaction is quenched with saturated aqueous NH4Cl solution (20 ml) and extracted with EtOAc (2 x 30 ml). The combined organic solution is washed with brine (30 ml), dried over MgSO4 and concentrated. Silica gel chromatography yields the desired product.
  • Step B: 1-(2-(Tert-butyldimethylsilyloxy)ethyl)-N-(7-chloro-6-(2-fluoro-4-iodophenylamino)-3-methyl-3a,7a-dihydro-3H-imidazo[4,5-b]pyridin-5-yl)cyclopropane-1-sulfonamide:
  • Figure imgb0134
  • To a solution of 3-(1-(2-(tert-butyldimethylsilyloxy)ethyl)cyclopropylsulfonyl)-8-chloro-1-(2-fluoro-4-iodophenyl)-5-methyl-3,4a,5,7a-tetrahydrodiimidazo[4,5-b:4',5'-e]pyridin-2(1H)-one (86 mg, 0.124 mmol) in THF (5 ml) is added potassium trimethylsilanolate (80 mg, 0.62 mmol). The resulting solution is stirred at room temperature for 24 h. The reaction is quenched with saturated aqueous NH4Cl solution (10 ml) and extracted with EtOAc (2 x 10 ml). The combined organic solution is washed with brine (10 ml), dried over MgSO4 and concentrated. Silica gel chromatography yields the desired product.
  • Step C: N-(7-chloro-6-(2-fluoro-4-iodophenylamino)-3-methyl-3a,7a-dihydro-3H-imidazo[4,5-b]pyridin-5-yl)-1-(2-hydroxyethyl)cyclopropane-1-sulfonamide:
  • Figure imgb0135
  • To a solution of 1-(2-(tert-butyldimethylsilyloxy)ethyl)-N-(7-chloro-6-(2-fluoro-4-iodophenylamino)-3-methyl-3a,7a-dihydro-3H-imidazo[4,5-b]pyridin-5-yl)cyclopropane-1-sulfonamide (40 mg, 0.060 mmol) in THF (2 ml) is added HCl (0.20 ml, 1.2 N in H2O, 0.24 mmol). After stirring for 1 h, the reaction is quenched with saturated aqueous NaHCO3 solution (3 ml) and extracted with EtOAc (2 x 10 ml). The combined organic solution is dried over Na2SO4 and concentrated. Silica gel chromatography yields the title compound.
  • Example 23 (not claimed): N-(7-(4-bromo-2-fluorophenylamino)imidazo[1,2-b]pyridazin-6-yl)cyclopropanesulfonamide: Step A: Preparation of 4,6-dichloro-pyridazine-3-carboxylic acid:
  • Figure imgb0136
  • To a solution of methyl 4,6-dichloro-pyridazine-3-carboxylate (6.01 g, 29.03 mmol, WO 2004/031174 ) in THF (29 ml) at 0 °C is added 29 ml of aqueous 1.0 M LiOH with stirring. The resulted mixture is stirred continuously at 0 °C for 40 min and monitored by TLC. The reaction mixture is diluted with water, and acidified to pH 1-2 with aqueous 1.0 M HCl. The crude product is suspended between the organic and aqueous layers. After filtration and washed with water and hexane, and dried under vacuum, the product is obtained.
  • Step B: 4-(4-Bromo-2-fluoro-phenylamino)-6-chloro-pyridazine-3-carboxylic acid:
  • Figure imgb0137
  • To the stirred solution of 4-bromo-2-fluorophenylamine (3.15 g, 16.58 mmol) in THF (50 ml) under argon at-78 °C is added dropwise LiHMDS (24.9 ml, 24.9 mmol, 1.0 M in hexane). After stirring for one hour at -78 °C, a solution of 4,6-dichloropyridazine-3-carboxylic acid (1.6 g, 8.29 mmol) in THF (10 ml) is added dropwise. The resulting mixture is allowed to warm to room temperature slowly and was stirred continuously for 18 hours. The reaction is quenched by addition of 5 ml of water, diluted with ethyl acetate, acidified with aqueous 1.0 M HCl to pH 1-2. The layers are separated and aqueous layer is extracted with ethyl acetate (30 ml x 3). The combined organic layers are washed with brine (30 ml x 3), dried over Na2SO4, and concentrated under reduced pressure. The residue is purified by flash column chromatography to give the product.
  • Step C: 4-(4-Bromo-2-fluoro-phenylamino)-6-chloro-pyridazine-3-carboxylic acid tert-butyl ester:
  • Figure imgb0138
  • To the stirred solution of 4-(4-bromo-2-fluoro-phenylamino)-6-chloro-pyridazine-3-carboxylic acid (516 gm, 1.49 mmol) in THF (10 ml) under argon at room temperature is added a solution of 2-tert-butyl-1,3-diisopropylisourea (1.49 g, 7.45 mmol) in THF (1.0 ml). The resulted mixture is refluxed for 6 hours. The reaction mixture is then cooled to room temperature and diluted with ethyl acetate. The organic layer is washed with 10% K2CO3 (20 ml x 2) and saturated NaCl (30 ml x 3), dried over Na2SO4 and concentrated in vacuum. The residue is dissolved in 30 ml of dichloromethane and the resulting white solid (urea byproduct) is filtered out. The filtrate is concentrated under reduced pressure. The residue is purified by flash column chromatography to provide the desired product.
  • Step D: 6-Azido-4-(4-Bromo-2-fluoro-phenylamino)-pyridazine-3-carboxylic acid tert-butyl ester:
  • Figure imgb0139
  • To a solution of 4-(4-bromo-2-fluoro-phenylamino)-6-chloro-pyridazine-3-carboxylic acid tert-butyl ester (300 mg, 0.74 mmol) in DMF (5 ml) under argon at room temperature is added sodium azide (96 mg, 1.48 mmol) with stirring. The reaction mixture is stirred at 50 °C for 6 hours. After cooling to room temperature, the reaction mixture is diluted with ethyl acetate and washed with water, saturated NaHCO3 (20 ml x 3) and saturated NaCl (30 ml x 3). The organic layer is dried over Na2SO4 and concentrated. The residue is purified by flash column chromatography to provide the desired product.
  • Step E: 6-Amino-4-(4-Bromo-2-fluoro-phenylamino)-6-chloro-pyridazine-3-carboxylic acid tert-butyl ester:
  • Figure imgb0140
  • To the stirred solution of 6-azido-4-(4-bromo-2-fluoro-phenylamino)-6-chloro-pyridazine-3-carboxylic acid tert-butyl ester (122 mg, 0.30 mmol) in 5 ml of mixed solvents (3 : 1 v/v, dichloromethane / acetic acid) is added zinc powder (98 mg, 1.5 mmol) at room temperature. After three hours, the reaction mixture is poured into ethyl acetate (50 ml). The organic layer is washed with water, saturated NaHCO3 (30 ml x 3) and saturated NaCl (30 ml x 3). The organic layer is dried over Na2SO4, and concentrated. The residue is purified by flash column chromatography to provide the desired product.
  • Step F: 7-(4-Bromo-2-fluoro-phenylamino)-imidazo[1,2-b]pyridazine-6-carboxylic acid:
  • Figure imgb0141
  • To a suspended solution of 6-amino-4-(4-bromo-2-fluoro-phenylamino)-6-chloro-pyridazine-3-carboxylic acid tert-butyl ester (61 mg, 0.16 mmol) in ethanol (5 ml) in a bomb reactor is added at room temperature chloroacetaldehyde (0.12 ml of 50% aqueous solution, 5.0 equivalents). The reaction mixture is sealed and stirred at 80 °C for two days, and then cooled to room temperature. The solvent is removed, and the residue is diluted with ethyl acetate (20 ml). The organic layer is washed with saturated NaCl (20 ml x 3), dried over Na2SO4, and concentrated. The residue is purified by flash column chromatography to provide the desired product.
  • Step G: 1-(4-Bromo-2-fluoro-phenyl)-1H,3H-1,3,4,4a,7-pentaaza-s-indacen-2-one:
  • Figure imgb0142
  • Synthesize according to example 21, step B from 7-(4-bromo-2-fluoro-phenylamino)-imidazo[1,2-b]pyridazine-6-carboxylic acid to afford the title compound.
  • Step H: 1-(4-Bromo-2-fluoro-phenyl)-3-(butane-2-sulfonyl)-1H,3H-1,3,4,4a,7-pentaaza-s-indacen-2-one:
  • Figure imgb0143
  • Synthesize according to example 21, step C from 1-(4-bromo-2-fluoro-phenyl)-1H,3H-1,3,4,4a,7-pentaaza-s-indacen-2-one to afford the title compound.
  • Step I: Cyclopropanesulfonic acid [7-(4-bromo-2-fluoro-phenylamino)-imidazo[1,2-b]pyridazin-6-yl]-amide:
  • Figure imgb0144
  • Synthesize according to example 4, step D from 1-(4-bromo-2-fluoro-phenyl)-3-(butane-2-sulfonyl)-1H,3H-1,3,4,4a,7-pentaaza-s-indacen-2-one to afford the title compound.
  • Example 24 (not claimed): N-(7-(4-bromo-2-fluorophenylamino)imidazo[1,2-b]pyridazin-6-yl)-1-(2-hydroxyethyl)cyclopropane-1-sulfonamide:
  • Figure imgb0145
  • Synthesize according to example 22, step A, B and C from 1-(4-bromo-2-fluoro-phenyl)-1H,3H-1,3,4,4a,7-pentaaza-s-indacen-2-one to afford the title compound.
  • Example 25 (not claimed): N-(7-fluoro-6-(2-fluoro-4-iodophenylamino)pyrazolo[1,5-a]pyrimidin-5-yl)cyclopropanesulfonamide:
  • Figure imgb0146
  • This compound is synthesized according to example 21 (step A through D) starting from 6-chloro-7-fluoropyrazolo[1,5-a]pyrimidine-5-carboxylic acid ( WO 2005/051906 ).
  • Example 26 (not claimed): N-(7-fluoro-6-(2-fluoro-4-iodophenylamino)pyrazolo[1,5-a]pyrimidin-5-yl)-1-(2-hydroxyethyl)cyclopropane-1-sulfonamide:
  • Figure imgb0147
  • This compound is synthesized according to example 22 (step A through C) starting from 3-(cyclopropylsulfonyl)-9-fluoro-1-(2-fluoro-4-iodophenyl)-1H-pyrazolo[1,5-a]purin-2(3H)-one (example 25).
  • Example 27 (not claimed): N-(6-(4-bromo-2-fluorophenylamino)-7-fluoro-3-methylisoxazolo[4,5-b]pyridin-5-yl)cyclopropanesulfonamide:
  • Figure imgb0148
  • This compound is synthesized according to example 4 (step B through D) starting from 6-(4-bromo-2-fluorophenylamino)-7-fluoro-3-methylisoxazolo[4,5-b]pyridine-5-carboxylic acid ( WO 2005/051906 ).
  • Example 28 (not claimed): N-(6-(4-bromo-2-fluorophenylamino)-7-fluoro-3-methylisoxazolo[4,5-b]pyridin-5-yl)-1-(2-hydroxyethyl)cyclopropane-1-sulfonamide:
  • Figure imgb0149
  • This compound is synthesized according to example 4 (step A through C) starting from 7-(4-bromo-2-fluorophenyl)-8-fluoro- 3-methyl-5H-imidazo[4,5-e]isoxazolo[4,5-b]pyridin-6(7H)-one (example 27).
  • Example 29 (not claimed): N-(6-(2-fluoro-4-iodophenylamino)-7-fluoro-2,3-dimethyl-2H-indazol-5-yl)cyclopropanesulfonamide: Step A: 7-fluoro-6-(2-fluoro-phenylamino)-3-methyl-2H-indazole-5-carboxylic acid methyl ester:
  • Figure imgb0150
  • To a solution of methyl 5-acetyl-2-(2-fluorophenylamino)-3,4-difluorobenzoate (500 mg, 1.55 mmol, from step D in example 3) in DMF (10 ml) was added hydrazine (0.06 mL, 1.85 mmol). After stirring at room temperature for 16 h, the reaction was quenched with NH4Cl solution and extracted with EtOAc. The organic layer was dried (MgSO4) and concentrated under reduced pressure to afford 740 mg (75%) of the title compound.
  • Step B: 7-fluoro-6-(2-fluoro-phenylamino)-2,3-dimethyl-2H-indazole-5-carboxylic acid methyl ester:
  • Figure imgb0151
  • To a solution of 7-fluoro-6-(2-fluoro-phenylamino)-3-methyl-2H-indazole-5-carboxylic acid methyl ester (146 mg, 0.46 mmol) in DMF (5 ml) were added potassium carbonate (95 mg, 069 mmol) and iodomethane (0.034 mL, 0.55 mmol). The reaction was stirred at room temp overnight, diluted with EtOAc and washed with H2O. The organic fraction was dried over MgSO4 and concentrated under reduced pressure. Silica gel chromatography (EtOAc:Hexanes = 75:25) yielded 40 mg (26%) of the title compound and 81 mg (53%) of 7-fluoro-6-(2-fluoro-phenylamino)-1,3-dimethyl-1H-indazole-5-carboxylic acid methyl ester.
  • Step C: 7-fluoro-6-(2-fluoro-4-iodophenylamino)-2,3-dimethyl-2H-indazole-5-carboxylic acid methyl ester:
  • Figure imgb0152
  • To a solution of 7-fluoro-6-(2-fluoro-phenylamino)-2,3-dimethyl-2H-indazole-5-carboxylic acid methyl ester (141 mg, 0.43 mmol) in DMF (5 ml) were added NIS (144 mg, 0.64 mmol) and TFA (0.20 mL), and the reaction was stirred at room temp for 4 h. The crude was diluted with EtOAc and washed with H2O. The organic fraction is dried over MgSO4 and concentrated under reduced pressure. Silica gel chromatography (EtOAc:Hexanes = 75:25) yielded 137 mg (70%) of the title compound as a yellow solid.
  • Step D: 7-fluoro-6-(2-fluoro-4-iodophenylamino)-2,3-dimethyl-2H-indazole-5-carboxylic acid:
  • Figure imgb0153
  • To a solution of 7-fluoro-6-(2-fluoro-4-iodophenylamino)-2,3-dimethyl-2H-indazole-5-carboxylic acid methyl ester (137 mg, 0.30 mmol) in a mixture of THF (10 ml) and H2O (5 ml) was added a solution of LiOH (1.50 mL, 1M in H2O, 1.50 mmol). After stirring at room temperature for 4 h, the reaction was concentrated, acidified with 1 M HCl solution, and extracted with EtOAc (2 x 20 ml). The organic solution was dried over MgSO4 and concentrated to give 126 mg (96%) the title compound as an orange solid.
  • Step E: 8-fluoro-7-(2-fluoro-4-iodophenyl)-2,3-dimethyl-2,7-dihydro-5H-imidazo[4,5-f]indazol-6-one:
  • Figure imgb0154
  • To a suspension of 7-fluoro-6-(2-fluoro-4-iodophenylamino]-2,3-dimethyl-2H-indazole-5-carboxylic acid (126 mg, 0.29 mmol) in toluene (10 ml) was added triethylamine (0.13 ml, 0.90 mmol) and diphenylphosphoryl azide (0.081 ml, 0.38 mmol). The reaction was heated at reflux for 4 h. The mixture was cooled to room temperature and concentrated to dryness. Silica gel chromatography (EtOAc:Hexane = 75:25) yields 51 mg (39%) of the title compound as a light yellow solid.
  • Step F: 5-cyclopropanesulfonyl-8-fluoro-7-(2-fluoro-4-iodophenyl)-2,3-dimethyl-2,7-dihydro-5H-imidazo[4,5-f]indazol-6-one:
  • Figure imgb0155
  • To a solution of 8-fluoro-7-(2-fluoro-4-iodophenyl)-2,3-dimethyl-2,7-dihydro-5H-imidazo[4,5-f]indazol-6-one (34 mg, 0.077 mmol) in THF (5 ml) at -78 °C was added LiHMDS (0.12 ml, 1 M in THF, 0.12mmol). The reaction mixture was stirred at -78 °C for 10 min and cyclopropylsulfonyl chloride (22 mg, 0.15mmol) was added to the mixture. The reaction was slowly warmed to room temperature and stirred at the temperature for 16 h. The reaction was quenched with saturated aqueous NH4Cl solution (10 ml) and extracted with EtOAc (2 x 10 ml). The combined organic solution was washed with brine (20 ml), dried over MgSO4 and concentrated. Silica gel chromatography (EtOAc:Hexanes = 25:75) yields 24 mg (57%) of the title compound as a yellow solid.
  • Step G: N-(6-(2-fluoro-4-iodophenylamino)-7-fluoro-2,3-dimethyl-3H-indazol-5-yl)cyclopropanesulfonamide:
  • Figure imgb0156
  • To a solution of 5-cyclopropanesulfonyl-8-fluoro-7-(2-fluoro-4-iodophenyl)-2,3-dimethyl-2,7-dihydro-5H-imidazo[4,5-f]indazol-6-one (24 mg, 0.044 mmol) in THF (3 ml) was added potassium trimethylsilanolate (11 mg, 0.088 mmol). The reaction was stirred at room temperature for 5 h, quenched with saturated aqueous NH4Cl solution (5 ml), and extracted with EtOAc (2 x 10 ml). The combined organic solution was washed with brine (15 ml), dried over MgSO4, and concentrated. Silica gel chromatography (EtOAc:Hexanes = 50:50) yielded 19 mg (83%) of the title compound. m/z = 517 [M-1]-. 1H NMR (DMSO-d 6, 400 MHz): δ 0.78 (m, 4H), 2.48 (m, 1H), 2.68 (s, 3H), 4.13 (s, 3H), 6.38 (m, 1H), 7.32 (d, J = 8 Hz, 1H), 7.41 (s, 1H), 7.59 (m, 2H), 9.23 (s, 1H).
  • Example 30 (not claimed): N-(6-(2-fluoro-4-iodophenylamino)-7-fluoro-1,3-dimethyl-1H-indazol-5-yl)cyclopropanesulfonamide:
  • Figure imgb0157
  • This compound was synthesized according to example 29 (step C through G) starting from 7-fluoro-6-(2-fluoro-phenylamino)-1,3-dimethyl-1H-indazole-5-carboxylic acid methyl ester (step B, example 29). m/z = 517 [M-1]-. 1H NMR (DMSO-d 6, 400 MHz): δ 0.71 (m, 2H), 0.79 (m, 2H), 2.49 (m, 1H), 2.47 (s, 3H), 4.05 (s, 3H), 6.49 (m, 1H), 7.26 (d, J = 8 Hz, 1H), 7.48 (s, 1H), 7.55 (s, 1H), 7.57 (s, 1H), 9.26 (s, 1H)
  • Example 31 (not claimed): N-(6-(2-fluoro-4-iodophenylamino)-7-fluoro-2,3-dimethyl-2H-indazol-5-yl)-1-(2-hydroxyethyl)cyclopropane-1-sulfonamide:
  • Figure imgb0158
  • This compound is synthesized according to example 4 (step A through C) starting from 8-fluoro-7-(2-fluoro-4-iodophenyl)-2,3-dimethyl-2,7-dihydro-5H-imidazo[4,5-f]indazol-6-one (example 29).
  • Example 32: N-(7-fluoro-6-(2-fluoro-4-iodophenylamino)-3-methylisoxazolo[4,3-b]pyridin-5-yl)cyclopropanesulfonamide: Step A: Methyl 7-fluoro-6-(2-fluorophenylamino)-3-methylisoxazolo[4,3-b]pyridine-5-carboxylate:
  • Figure imgb0159
  • To a solution of methyl 6-acetyl-5-chloro-4-fluoro-3-(2-fluorophenylamino)picolinate ( WO 2005/051906 ) (2 g, 5 mmole) in a mixture of acetone (36 ml) and H2O (12 ml) is added NaN3 (487 mg, 7.5 mmol). The reaction is heated at 65 °C for 16 h and cooled to room temperature. Acetone is concentrated and the resulting mixture is extracted with EtOAc (2 x 50 ml). The combined organic solution is washed with brine (50 ml), dried over MgSO4 and concentrated to give a yellow solid. To the solid is added H2O (24 ml) and the reaction is heated at reflux for 3 h. The reaction is cooled to room temperature, filtered, and washed with H2O. The resulting solid is dried under high vacuum with P2O5 at 65°C for 24 h to give the title compound.
  • Step B: Methyl 7-fluoro-6-(2-fluoro-4-iodophenylamino)-3-methylisoxazolo[4,3-b]pyridine-5-carboxylate:
  • Figure imgb0160
  • To a suspension of methyl 7-fluoro-6-(2-fluorophenylamino)-3-methylisoxazolo[4,3-b]pyridine-5-carboxylate (1.35 g, 4.25 mmol) in DMF (40 ml) is added N-iodosuccinimide (1.43 g, 6.37 mmol) and TFA (2.5 ml). After stirring at room temperature for 4 h, the reaction is quenched with saturated aqueous NH4Cl solution (100 ml) and extracted with EtOAc (2 x 100 ml). The combined organic solution is washed with brine (100 ml), dried over MgSO4 and concentrated. Silica gel chromatography yields the title compound.
  • Step C: 7-Fluoro-6-(2-fluoro-4-iodophenylamino)-3-methylisoxazolo[4,3-b]pyridine-5-carboxylic acid:
  • Figure imgb0161
  • To a solution of methyl 7-fluoro-6-(2-fluoro-4-iodophenylamino)-3-methylisoxazolo[4,3-b]pyridine-5-carboxylate (250 mg, 0.56 mmol) in a mixture of THF (10 ml) and H2O (5 ml) is added a solution of LiOH (67 mg, 2.8 mmol) in H2O (2.4 ml). After stirring at room temperature for 4 h, the reaction is concentrated, acidified with 1 M HCl solution, and extracted with EtOAc (2 x 20 ml). The organic solution is dried over MgSO4 and concentrated to give the title compound.
  • Step D: 8-Fluoro-7-(2-fluoro-4-iodophenyl)-3-methyl-5H-imidazo[4,5-b]isoxazolo[3,4-e]pyridin-6(7H)-one:
  • Figure imgb0162
  • To a suspension of 7-fluoro-6-(2-fluoro-4-iodophenylamino)-3-methylisoxazolo[4,3-b]pyridine-5-carboxylic acid (241 mg, 0.56 mmol) in toluene (10 ml) is added triethylamine (0.23 ml, 1.68 mmol) and diphenylphosphoryl azide (0.26 ml, 1.21 mmol). The reaction is heated at reflux for 4 h. The mixture is cooled to room temperature and concentrated to dryness. Silica gel chromatography yields the title compound.
  • Step E: 5-(Cyclopropylsulfonyl)-8-fluoro-7-(2-fluoro-4-iodophenyl)-3-methyl-5H-imidazo[4,5-b]isoxazolo[3,4-e]pyridin-6(7H)-one:
  • Figure imgb0163
  • To a solution of 8-fluoro-7-(2-fluoro-4-iodophenyl)-3-methyl-5H-imidazo[4,5-b]isoxazolo[3,4-e]pyridin-6(7H)-one (143 mg, 0.33 mmol) in THF (10 ml) at -78 °C is added LiHMDS (0.38 ml, 1 M in THF, 0.38 mmol). The reaction mixture is stirred at -78 °C for 10 min and cyclopropylsulfonyl chloride (69 mg, 0.50 mmol) is added to the mixture. The reaction is slowly warmed to room temperature and stirred at the temperature for 16 h. The reaction is quenched with saturated aqueous NH4Cl solution (20 ml) and extracted with EtOAc (2 x 20 ml). The combined organic solution is washed with brine (20 ml), dried over MgSO4 and concentrated. Silica gel chromatography yields the title compound.
  • Step F: N-(7-fluoro-6-(2-fluoro-4-iodophenylamino)-3-methylisoxazolo[4,3-b]pyridin-5-yl)cyclopropanesulfonamide:
  • Figure imgb0164
  • To a solution of 3-(cyclopropanesulfonyl)-8-fluoro-1-(2-fluoro-4-iodophenyl)-5-methylbenzimidazolo[6,5-d][c]isoxazole (131 mg, 0.25 mmol) in THF (15 ml) is added potassium trimethylsilanolate (95 mg, 0.75 mmol). The reaction is stirred at room temperature for 16 h, quenched with saturated aqueous NH4Cl solution (20 ml), and extracted with EtOAc (2 x 30 ml). The combined organic solution is washed with brine (30 ml), dried over MgSO4, and concentrated. Silica gel chromatography yields the title compound.
  • Example 33: N-(7-fluoro-6-(2-fluoro-4-iodophenylamino)-3-methylisoxazolo[4,3-b]pyridin-5-yl)-1-(2-hydroxyethyl)cyclopropane-1-sulfonamide:
  • Figure imgb0165
  • This compound is synthesized according to example 4 (step A through C) starting from 8-fluoro-7-(2-fluoro-4-iodophenyl)-3-methyl-5H-imidazo[4,5-b]isoxazolo[3,4-e]pyridin-6(7H)-one (example 32).
  • Example 34 (not claimed): N-(7-(2-fluoro-4-iodophenylamino)-8-fluoro-4-methylquinazolin-6-yl)cyclopropanesulfonamide: Step A: 5-Bromo-2,3,4-trifluorobenzoic acid:
  • Figure imgb0166
  • To a solution of iPr2NH (56.8 mmol, 8ml) in THF (50 ml) at -78 °C was added dropwise a solution of nBuLi (1.6M in hexane, 56.8 ml, 35.5 mmol the dry ice bath was removed and the mixture was stirred at 0 °C for ∼ 15 min. The mixture is transferred via cannula to a solution of 1-bromo-2,3,4-trifluorobenzene (47.4 mmol, 10 g) in THF (50 ml) cooled at -78 °C. The orange solution was stirred at -78 °C for 1 h, then transferred, via cannula, to freshly grinded dry CO2. The reaction was warmed up to room temperature and stirred overnight. The reaction mixture was cooled at 0 °C, quenched with 10% HCl (300 ml), extracted with ether (100 ml x 3), dry over MgSO4 and concentrated under reduced pressure to obtain (10.6 g, 88%) of the title compound.
  • Step B: 5-Bromo-2-(2-fluorophenylamino)-3,4-difluorobenzoic acid:
  • Figure imgb0167
  • To a solution of 3-fluoro-aniline (177 mmol, 17.0 ml) in THF (80 mL) at -78 °C was added LiHMDS (265 mmol, 265 ml) and stirred for 30 min. To the reaction was added a solution of 5-bromo-2,3,4-trifluorobenzoic acid (78.4 mmol, 20.1 g) in THF (60 ml) at -78 °C. The reaction was warmed to room temperature and stirred at the temperature over night. The reaction is concentrated and slowly 10% HCl (180 ml) is added. The mixture was extracted with ethyl acetate (100 ml x 3), dried over MgSO4 and concentrated under reduced pressure. The crude material was taken in CH2Cl2 (∼250 ml) and heated up to 45 °C for 10 min. Purification by trituration with CH2Cl2 gave 11 g (72%) of the desired product as a light yellow solid.
  • Step C: Methyl 5-bromo-2-(2-fluorophenylamino)-3,4-difluorobenzoate:
  • Figure imgb0168
  • To a solution of 5-bromo-2-(2-fluorophenylamino)-3,4-difluorobenzoic acid (30.3 mmol, 11 g) in THF:MeOH (60:20), cooled to 0 °C, was added dropwise a solution of 2M TMSCHN2 in hexane (39.39 mmol, 19.7 ml). After the addition is completed, the reaction mixture was warmed to room temperature and stirred for an additional 4 hours. The crude reaction was quenched with acetic acid and extracted with EtOAc. The organic layers are combined, washed with H2O, dry over MgSO4 and concentrated under reduced pressure. The crude material was dried under vacuum and yields to 11g (96%) of the title product.
  • Step D: Methyl 2-(2-fluorophenylamino)-3,4-difluoro-5-((trimethylsilyl)ethynyl)benzoate:
  • Figure imgb0169
  • To an oven dried flask charged with a solution of methyl 5-bromo-2-(2-fluorophenylamino)-3,4-difluorobenzoate (21.68mmol, 8.1g) in THF (116ml), was added CuI (2.16 mmol, 0.4 g) and iPr2NH (43.36 mmol, 6.1 ml). After addition is completed, reaction was purged with Argon and Pd(PPh3)2Cl2 (3.73 mmol, 2.62 g) and TMS-acetylene (26.88 mmol, 3.7 ml) are added. The reaction mixture was stirred at room temperature for 16 h, concentrated and taken up in EtOAc. The organic fraction was washed with sat NH4Cl, brine, dried over MgSO4 and conc. The crude was purified by flash chromatography using 5% to 20% EtOAc in Hexane to afford the desired product 6.96 g (85%).
  • Step E: Methyl 5-acetyl-2-(2-fluorophenylamino)-3,4-difluorobenzoate :
  • Figure imgb0170
  • A mixture of methyl 2-(2-fluorophenylamino)-3,4-difluoro-5-((trimethylsilyl)ethynyl)benzoate (9 mmol, 3.5 g), HgSO4 (9 mmol, 2.67 g), and conc. H2SO4 (18 mmol, 0.96 ml) in acetone:H2O (70:12 ml) was refluxed at 65 °C overnight. The mixture was concentrated, diluted with EtOAc, and washed with water and brine. The organic fractions are combined, dried over MgSO4 and concentrated under reduced pressure. The crude material (brown solid) was triturated with MeOH and the precipitate was filtered, washed with additional MeOH and dried to yield the desired product in 60% (1.78 g).
  • Step F: Methyl 5-acetyl-2-(2-fluoro-4-iodophenylamino)-3,4-difluorobenzoate :
  • Figure imgb0171
  • To a solution of methyl 5-acetyl-2-(2-fluorophenylamino)-3,4-difluorobenzoate (0.47 mmol, 0.16 g) in DMF (10 ml) was added NIS (0.56 mmol, 0.13 g) and TFA (0.10 ml), and the reaction was stirred at room temp overnight. The crude was diluted with EtOAc and washed with H2O. The organic fraction was dried over MgSO4 and concentrated under reduced pressure to yield the desired product in 90% (0.23 g).
  • Step G: Methyl 7-(2-fluoro-4-iodophenylamino)-8-fluoro-4-methylquinazoline-6-carboxylate:
  • Figure imgb0172
  • A mixture of methyl 5-acetyl-2-(2-fluoro-4-iodophenylamino)-3,4-difluorobenzoate (230 mg, 0.55 mmol) and formamide acetate (68 mg, 0.66 mmol) in DMA (5 ml) was heated at 95 °C for 6 hours. The reaction mixture was diluted with EtOAc and washed with water and brine. The organic layer was dried (MgSO4) and concentrated under reduced pressure to afford 205 mg (88%) of the title compound.
  • Step H: 7-(2-Fluoro-4-iodophenylamino)-8-fluoro-4-methylquinazoline-6-carboxylic acid:
  • Figure imgb0173
  • To a solution of methyl 7-(2-fluoro-4-iodophenylamino)-8-fluoro-4-methylquinazoline-6-carboxylate (205 mg, 0.48 mmol) in a mixture of THF (10 ml) and H2O (5 ml) was added a solution of LiOH (57 mg, 2.4 mmol) in H2O (2.4 ml). After stirring at room temperature for 4 h, the reaction was concentrated, acidified with 1 M HCl solution, and extracted with EtOAc (2 x 20 ml). The organic solution was dried over MgSO4 and concentrated to give 190 mg (96%) the title compound as an orange solid.
  • Step I: 3-(2-Fluoro-4-iodophenyl)-4-fluoro-8-methyl-1H-imidazo[4,5-g]quinazolin-2(3H)-one:
  • Figure imgb0174
  • To a suspension of 7-(2-fluoro-4-iodophenylamino)-8-fluoro-4-methylquinazoline-6-carboxylic acid (190 mg, 0.47 mmol) in toluene (10 ml) was added triethylamine (0.19 ml, 1.41 mmol) and diphenylphosphoryl azide (0.20 ml, 0.94 mmol). The reaction was heated at reflux for 4 h. The mixture was cooled to room temperature and concentrated to dryness. Silica gel chromatography (EtOAc:Hexane = 75:25) yields 143 mg (75%) of the title compound as a tan solid.
  • Step J: 3-(2-Fluoro-4-iodophenyl)-1-(cyclopropylsulfonyl)-4-fluoro-8-methyl-1H-imidazo[4,5-g]quinazolin-2(3H)-one:
  • Figure imgb0175
  • To a solution of 3-(2-fluoro-4-iodophenyl)-4-fluoro-8-methyl-1H-imidazo[4,5-g]quinazolin-2(3H)-one (50 mg, 0.12 mmol) in THF (10 ml) at -78 °C was added LiHMDS (0.16 ml, 1 M in THF, 0.16 mmol). The reaction mixture was stirred at -78 °C for 10 min and cyclopropylsulfonyl chloride (25 mg, 0.18 mmol) was added to the mixture. The reaction was slowly warmed to room temperature and stirred at the temperature for 16 h. The reaction was quenched with saturated aqueous NH4Cl solution (10 ml) and extracted with EtOAc (2 x 10 ml). The combined organic solution was washed with brine (20 ml), dried over MgSO4 and concentrated. Silica gel chromatography (EtOAc:Hexanes = 25:75) yields 46 mg (75%) of the title compound as a yellow solid.
  • Step K: N-(7-(2-Fluoro-4-iodophenylamino)-8-fluoro-4-methylquinazolin-6-yl)cyclopropanesulfonamide:
  • Figure imgb0176
  • To a solution of 3-(2-fluoro-4-iodophenyl)-1-(cyclopropylsulfonyl)-4-fluoro-8-methyl-1H-imidazo[4,5-g]quinazolin-2(3H)-one (46 mg, 0.09 mmol) in THF (5 ml) was added potassium trimethylsilanolate (35 mg, 0.27 mmol). The reaction was stirred at room temperature for 16 h, quenched with saturated aqueous NH4Cl solution (5 ml), and extracted with EtOAc (2 x 10 ml). The combined organic solution was washed with brine
  • (15 ml), dried over MgSO4, and concentrated. Silica gel chromatography (EtOAc:Hexanes = 50:50) yielded 39 mg (90%) of the title compound as a light yellow solid. m/z = 515 [M-1]-. 1H NMR (DMSO-d 6, 400 MHz): δ 0.85 (m, 4H), 2.75 (m, 1H), 2.87 (s, 3H), 6.61 (m, 1H), 7.36 (d, J = 8 Hz, 1H), 7.67 (d, J= 11 Hz, 1H), 8.02 (s, 2H), 9.07 (s, 1H), 9.78 (s, 1H).
  • Example 35 (not claimed): N-(7-(2-fluoro-4-iodophenylamino)-8-fluoro-4-methylquinazolin-6-yl)-1-(2-hydroxyethyl)cyclopropane-1-sulfonamide: Step A: 3-(2-Fluoro-4-iodophenyl)-1-(1-(2-(tert-butyldimethylsilyloxy)ethyl)cyclopropylsulfonyl)-4-fluoro-8-methyl-1H-imidazo[4,5-g]quinazolin-2(3H)-one:
  • Figure imgb0177
  • To a solution of 3-(2-fluoro-4-iodophenyl)-4-fluoro-8-methyl-1H-imidazo[4,5-g]quinazolin-2(3H)-one (example 34, step H) (90 mg, 0.22 mmol) in THF (5 ml) at -78 °C is added LiHMDS (0.29 ml, 1 M in THF, 0.29 mmol). The reaction mixture is stirred at -78 °C for 30 min and 1-(2-(tert-butyldimethylsilyloxy)ethyl)cyclopropane-1-sulfonyl chloride (99 mg, 0.33 mmol) is added to the mixture. The reaction is slowly warmed to room temperature and stirred at the temperature for 16 h. The reaction is quenched with saturated aqueous NH4Cl solution (20 ml) and extracted with EtOAc (2 x 30 ml). The combined organic solution is washed with brine (30 ml), dried over MgSO4 and concentrated. Silica gel chromatography yields the desired product.
  • Step B: N-(7-(2-Fluoro-4-iodophenylamino)-8-fluoro-4-methylquinazolin-6-yl)-1-(2-(tert-butyldimethylsilyloxy)ethyl)cyclopropane-1-sulfonamide:
  • Figure imgb0178
  • To a solution of 3-(2-fluoro-4-iodophenyl)-1-(1-(2-(tert-butyldimethylsilyloxy)ethyl)cyclopropylsulfonyl)-4-fluoro-8-methyl-1H-imidazo[4,5-g]quinazolin-2(3H)-one (66 mg, 0.1 mmol) in THF (5 ml) is added potassium trimethylsilanolate (65 mg, 0.5 mmol). The resulting solution is stirred at room temperature for 24 h. The reaction is quenched with saturated aqueous NH4Cl solution (10 ml) and extracted with EtOAc (2 x 10 ml). The combined organic solution is washed with brine (10 ml), dried over MgSO4 and concentrated. Silica gel chromatography yields the desired product.
  • Step C: N-(7-(2-Fluoro-4-iodophenylamino)-8-fluoro-4-methylquinazolin-6-yl)-1-(2-hydroxyethyl)cyclopropane-1-sulfonamide:
  • Figure imgb0179
  • To a solution of N-(7-(2-fluoro-4-iodophenylamino)-8-fluoro-4-methylquinazolin-6-yl)-1-(2-(tert-butyldimethylsilyloxy)ethyl)cyclopropane-1-sulfonamide (36 mg, 0.056 mmol) in THF (2 ml) is added HCl (0.18 ml, 1.2 N in H2O, 0.22 mmol). After stirring for 1 h, the reaction is quenched with saturated aqueous NaHCO3 solution (3 ml) and extracted with EtOAc (2 x 10 ml). The combined organic solution is dried over Na2SO4 and concentrated. Silica gel chromatography yields the title compound.
  • Example 36 (not claimed): N-(7-(2,4-dichlorophenylamino)-8-fluoro-4-methylcinnolin-6-yl)cyclopropanesulfonamide:
  • Figure imgb0180
  • This compound is synthesized according to example 34 starting from 7-(4-bromo-2-chlorophenylamino)-8-fluoro-4-methylcinnoline-6-carboxylic acid ( WO 2005/051302 ).
  • Example 37 (not claimed): N-(7-(2,4-dichlorophenylamino)-8-fluoro-4-methylcinnolin-6-yl)-1-(2-hydroxyethyl)cyclopropane-1-sulfonamide:
  • Figure imgb0181
  • This compound is synthesized according to example 35 (step A through C) starting from 8-(2,4-dichlorophenyl)-9-fluoro-4-methyl-6H-imidazo[4,5-g]cinnolin-7(8H)-one (example 36).
  • Example 38 (not claimed): N-(7-(4-bromo-2-fluorophenylamino)-8-fluoro-4-methylquinolin-6-yl)cyclopropanesulfonamide:
  • Figure imgb0182
  • This compound is synthesized according to example 34 starting from 7-(4-bromo-2-fluorophenylamino)-8-fluoro-4-methylquinoline-6-carboxylic acid ( WO 2005/051302 ).
  • Example 39 (not claimed): N-(7-(4-bromo-2-fluorophenylamino)-8-fluoro-4-methylquinolin-6-yl)-1-(2-hydroxyethyl)cyclopropane-1-sulfonamide:
  • Figure imgb0183
  • This compound is synthesized according to example 35 (step A through C) starting from 3-(4-bromo-2-fluorophenyl)-4-fluoro-8-methyl-1H-imidazo[4,5-g]quinolin-2(3H)-one (example 38).
  • Example 40: Biological activity Generation of IC50 Data
  • Materials and preparation of reagents: Human GST-MEK1 and the constitutively 5 active allele GST-MEK1CA (harboring the mutations Ser218Asp and Ser222Asp) can be subcloned into the yeast expression vector pGEM4Z (Promega, Madison, WI) from the wild type human MEK1 cDNA. GST-MEK1CA was expressed in Escherichia coli and can be partially purified using Glutathione Sepharose 4B affinity resin (Amersham Pharmacia Biotech, Piscataway, NJ). The ERK2 allele can be subcloned from MAPK2/Erk2 cDNA (wild type) in 10 pUSEamp (Upstate Biotechnology, Inc., Waltham, MA) into the vector pET21a (Novagen, Madison, WI) resulting in an N-terminal histidine-tagged mouse ERK2 allele. ERK2 can be expressed and purified to homogeneity [Zhang, 1993 #33]. Myelin basic protein (MBP) can be purchased from Gibco BRL (Rockville, MD). EasyTides adenosine 5'-triphosphate (ATP) ([γ-33P]) (NEN Perkin Elmer, Wellesley, MA) is the source of radiolabel for all kinase reactions. Activated Raf-1 (truncated) and activated MAPKinase 2/ERK2 can be purchased from Upstate, Inc. (Lake Placid, NY). 4-20% Criterion Precast gels can be purchased from Bio-Rad (Hercules, CA).
  • Determination of enzymatic activity: Compounds are diluted from dimethylsulfoxide (DMSO) stocks into 1xHMNDE (20 mM HEPES pH 7.2, 1 mM MgCl2, 100 mM NaCl, 1.25 mM DTT, 0.2 mM EDTA). A typical 25-microliter assay contained 0.002 nanomoles MEK1CA, 0.02 nanomoles ERK2, 0.25 nanomoles MBP, 0.25 nanomoles unlabeled ATP, and 0.1 µCi [γ33P] ATP. The screening assay essentially comprises four additions. Five µl of diluted compound are dispensed to 96-well assay plates. Ten µl of 2.5x enzyme cocktail (MEK1CA and ERK2 only) are then added to each well followed by a preincubation for 30 minutes at ambient temperature. Ten µl of 2.5x substrate cocktail (labeled and unlabeled ATP plus MBP) are then added, followed by incubation for 60 minutes at ambient temperature. Finally, 100 µl of 10% trichloroacetic acid (TCA) are added and incubated for 30 minutes at room temperature to halt the reaction and precipitate radiolabeled protein products. Reaction products are harvested on glass fiber 96 well filter plates prewetted with water and 1% pyrophosphate. The filter plate is then washed 5 times with water. Water is displaced by absolute ethanol and the plate is allowed to air dry for 30 minutes at room temperature. A back seal is applied manually and 40 µl of scintillation cocktail are dispensed to each well. A top seal is applied and the plate is counted in the TopCount for two seconds per well.
  • A truncated version of MEK that requires activation by Raf kinase can be used.
  • Generation of EC50 Data
  • Effects of compounds in the cell can be determined by Western blotting for phosphorylated ERK. MDA-MB-231 breast cancer cells are plated in a 48 well plate at 20,000 cells per well and are grown in a 37° humidified CO2 incubator. The following day, the growth media (DMEM + 10% fetal bovine serum) is removed and replaced with starve media (DMEM + 0.1% fetal bovine serum). Cells are incubated in the starve media for sixteen hours and then treated with a range of compound concentrations for thirty minutes.
  • After incubation with compound, cells are stimulated with 100ng/ml EGF for five minutes. The cells are then lysed and analyzed by Western blot using a monoclonal antibody raised to phosphorylated ERK. The signal is amplified using a secondary antibody conjugated to a near -IR dye and detected on a Licor Odyssey scanner. The intensity of signal is quantitated and this data was used to generate dose response curves and EC50 calculations.
    Example Structure IC50 (nM) EC50 (nM)
    2
    Figure imgb0184
    C B
    3
    Figure imgb0185
    C B
    4
    Figure imgb0186
    C B
    5
    Figure imgb0187
    B A
    6
    Figure imgb0188
    D B
    7
    Figure imgb0189
    C C
    9
    Figure imgb0190
    D C
    10
    Figure imgb0191
    D C
    12
    Figure imgb0192
    C B
    14
    Figure imgb0193
    C A
    16
    Figure imgb0194
    C C
    17
    Figure imgb0195
    D C
    18
    Figure imgb0196
    E C
    VALUE    RANGE
    A       0.1-1
    B       1-10
    C       10-100
    D       100-1000
    E       1000-10,000

Claims (19)

  1. A compound of formula (I):
    Figure imgb0197
    Figure imgb0198
    Figure imgb0199
    Figure imgb0200
    wherein
    G is R1a, R1b, R1c, R1d, R1e, Ar1, Ar2 or Ar3;
    Ro, R1 and R2 are independently selected from H, halogen, cyano, cyanomethyl, nitro, difluoromethoxy, difluoromethoxy, trifluoromethyl, azido, C1-C6 alkyl, C1-C4 alkoxy, C3-C6 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl; wherein
    said C1-C6 alkyl, C3-C6 cycloalkyl, C2-C6 alkenyl and C2-C6 alkynyl groups are optionally substituted with 1-3 substituents selected independently from halogen, OH, CN, cyanomethyl, nitro, phenyl, difluoromethoxy, difluoromethoxy, and trifluoromethyl;
    said C1-C6 alkyl and C1-C4 alkoxy groups are optionally substituted with OCH3 or OCH2CH3;
    X is F, Cl or methyl;
    Y is I, Br, Cl, CF3, C1-C3 alkyl, C2-C3 alkenyl, C2-C3 alkynyl, cyclopropyl, phenyl, pyridyl, pyrazolyl, OMe, OEt, or SMe, wherein
    all said methyl, ethyl, C1-C3 alkyl, and cyclopropyl groups of X and Y are optionally substituted with OH;
    all said phenyl, pyridyl, pyrazolyl groups of Y are optionally substituted with halogen, acetyl, methyl, and trifluoromethyl; and
    all said methyl groups of X and Y are optionally substituted with one, two, or three F atoms;
    Z is H, methyl, Cl or F;
    and wherein
    R1a is methyl, cyclopropoxy or C1- C4 alkoxy; wherein
    the methyl is optionally substituted with OH, 1-3 fluorine atoms or 1-3 chlorine atoms;
    the C1- C4 alkyl moieties of said C1- C4 alkoxy are optionally substituted with one hydroxy or methoxy group; and
    all C2- C4 alkyl groups within said C1- C4 alkoxy are optionally further substituted with a second OH group;
    R1b is CH(CH3)-C1-3 alkyl or C3-C6 cycloalkyl, said CH3, alkyl, and cycloalkyl groups optionally substituted with 1-3 substituents selected independently from F, Cl, Br, I, OH, C1-C4 alkoxy and CN.
    R1c is (CH2)nOmR', where
    m is 0 or 1; wherein
    when m is 1, n is 2 or 3, and
    when m is 0, n is 1 or 2;
    R' is C1-C6 alkyl, optionally substituted with 1-3 substituents selected independently from F, Cl, OH, OCH3, OCH2CH3, and C3-C6 cycloalkyl;
    R1d is C(A')(A")(B)- wherein
    B, A', and A" are, independently, H or C1-4 alkyl, optionally substituted with one or two OH groups or halogen atoms, or
    A' and A", together with the carbon atom to which they are attached, form a 3- to 6- member saturated ring, said ring optionally containing one or two heteroatoms selected, independently, from O, N, and S and optionally substituted with one or two groups selected independently from methyl, ethyl, and halo;
    R1e is benzyl or 2-phenyl ethyl, in which the phenyl group is optionally substituted
    Figure imgb0201
    where
    q is 1 or 2;
    R8 and R9 are, independently, H, F, Cl, Br, CH3, CH2F, CHF2, CF3, OCH3, OCH2F, OCHF2, OCF3, ethyl, n-propyl, isopropyl, cyclopropyl, isobutyl, sec-butyl, tert-butyl, and methylsulfonyl;
    R10 is H, F, Cl, Br, CH3, CH2F, CHF2, CF3, OCH3, OCH2F, OCHF2, OCF3, ethyl, n-propyl, isopropyl, cyclopropyl, isobutyl, sec-butyl, tert-butyl, and methylsulfonyl, nitro, acetamido, amidinyl, cyano, carbamoyl, methylcarbamoyl, dimethylcarbamoyl, 1,3,4-oxadiazol-2-yl, 5-methyl-1,3,4-5 oxadiazolyl, 1,3,4-thiadiazolyl, 5-methyl-1,3,4-thiadiazol-1H-tetrazolyl, N-morpholinyl carbonylamino, N-morpholinylsulfonyl or N-pyrrolidinylcarbonylamino;
    R11 and R12 are, independently, H, F, Cl, or methyl;
    Ar1 is
    Figure imgb0202
    where
    W and V are, independently, N, CR1 or CR2;
    R8, R9 and R10 are, independently, H, F, Cl, Br, CH3, CH2F, CHF2, CF3, OCH3, OCH2F, OCHF2, OCF3, ethyl, n-propyl, isopropyl, cyclopropyl, isobutyl, sec-butyl, tert-butyl, and methylsulfonyl, and R10 may also be nitro, acetamido, amidinyl, cyano, carbamoyl, methylcarbamoyl, dimethylcarbamoyl, 1,3,4-oxadiazol-2-yl, 5-methyl-1,3,4-oxadiazol, 1,3,4-thiadiazol, 5-methyl-1,3,4-thiadiazol 1H-tetrazolyl, N-morpholinylcarbonylamino, N-morpholinylsulfonyl and N-pyrrolidinylcarbonylamino;
    R11 and R12 are, independently, H, F, Cl or methyl;
    Ar2 is
    Figure imgb0203
    where
    the dashed line represents a double bond which may be located formally either between V and the carbon between W and V, or between W and the carbon between W and V;
    W is -S-, -O- or -N =, wherein
    when W is -O- or -S-, V is -CH=, -CCl= or -N =; and
    when W is -N =, V is CH= or -NCH3-;
    R13 and R14 are, independently, H, methoxycarbonyl, methylcarbamoyl, acetamido, acetyl, methyl, ethyl, trifluoromethyl or halogen;
    Ar3 is
    Figure imgb0204
    where
    W is -NH-, -NCH3- or -O-; and
    R13 and R14 are, independently, H, F, Cl, or methyl.
  2. The compound of claim 1, where
    X is F, Cl, or CH3;
    Y is I, Br, Cl, CF3, or C1-C3 alkyl; and
    Z is H or F.
  3. The compound of claim 1, where
    Ro is F, Cl, C1-C4 alkyl or C1-C4 alkoxy, said C1-C4 alkyl group and the C1-C4 alkyl moiety of said C1-C4 alkoxy group optionally substituted with F, Cl, OCH3, or OCH2CH3.
  4. The compound of claim 2, where
    Ro is H, F, Cl, C1-C4 alkyl, methoxy, ethoxy, or 2-methoxy-ethoxy.
  5. The compound of claim 1, where G is R1d.
  6. The compound of claim 5, where
    Ro is either :
    - fluoro, chloro, methyl, ethyl, propyl, isopropyl, sec-butyl, iso-butyl, tert-butyl, cyclopropyl, cyclobutyl, fluoromethyl, methoxy, fluoromethoxy, methylamino or dimethylamino; when
    X is F, Cl, CH3, or mono-, di- or trifluoromethyl;
    or :
    - F, Cl, methyl, ethyl, methoxy, ethoxy, or 2-methoxy-ethoxy; when
    X is F, Cl, or CH3;
    or :
    - H; when
    X is F, Cl, CH3, or mono-, di- or trifluoromethyl;
    and
    Y is I, Br, Cl, or mono-, di- or tri- fluoromethyl; and
    Z is H or F.
  7. The compound of claim 5, where C(A')(A") is C1-C6 cycloalkyl.
  8. The compound of claim 7, where B is H.
  9. The compound of claim 8, where C(A')(A") is cyclopropyl.
  10. The compound of claim 7, where B is methyl, optionally substituted with one OH group, or C2-C4 alkyl, optionally substituted with one or two OH groups.
  11. The compound of claim 10, where C(A')(A") is cyclopropyl.
  12. The compound of claim 11, where B is methyl, ethyl, 2-hydroxyethyl, n-propyl, 3-5 hydroxypropyl, 2,3-dihydroxypropyl, 3,4-dihydroxybutyl, isopropyl, 1-methyl-2-hydroxyethyl, n-butyl, sec-butyl, isobutyl, or 2-hydroxymethyl-3-hydroxy propyl.
  13. The compound of claim 12, where B is 2,3-dihydroxypropyl or 3,4-dihydroxybutyl.
  14. The compound of claim 13, in which the chiral carbon in B is in the R configuration.
  15. A composition comprising a compound according to claim 14, which is substantially free of the S isomer.
  16. A compound of claim 1, selected from the group of compounds consisting of:
    Figure imgb0205
    Figure imgb0206
    and
    Figure imgb0207
  17. A pharmaceutical composition comprising a pharmaceutically effective amount of a compound of formula I according to claim 1, or a pharmaceutically acceptable salt, ester, solvate or hydrate thereof, and a pharmaceutically acceptable carrier.
  18. Use of a compound of formula I according to claim 1, or a pharmaceutically acceptable salt, ester, solvate or hydrate thereof, for the preparation of a medicament for the treatment of a disorder or condition which is modulated by the MEK cascade.
  19. Use of a compound of formula I according to claim 1, or a pharmaceutically acceptable salt, ester, solvate or hydrate thereof, for the preparation of a medicament for the treatment of a hyperproliferative disorder.
EP08727965.9A 2007-01-19 2008-01-18 Inhibitors of mek Active EP2121620B1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US88584907P 2007-01-19 2007-01-19
PCT/US2008/051518 WO2008089459A1 (en) 2007-01-19 2008-01-18 Inhibitors of mek

Publications (2)

Publication Number Publication Date
EP2121620A1 EP2121620A1 (en) 2009-11-25
EP2121620B1 true EP2121620B1 (en) 2015-06-17

Family

ID=39333043

Family Applications (1)

Application Number Title Priority Date Filing Date
EP08727965.9A Active EP2121620B1 (en) 2007-01-19 2008-01-18 Inhibitors of mek

Country Status (19)

Country Link
US (2) US7820664B2 (en)
EP (1) EP2121620B1 (en)
JP (1) JP5491199B2 (en)
KR (1) KR20090111847A (en)
CN (1) CN101663279A (en)
AU (1) AU2008206045A1 (en)
BR (1) BRPI0806898A2 (en)
CA (1) CA2675358C (en)
CO (1) CO6210813A2 (en)
CR (1) CR10939A (en)
DO (1) DOP2009000181A (en)
EA (1) EA200900959A1 (en)
EC (1) ECSP099525A (en)
ES (1) ES2547303T3 (en)
IL (1) IL199727A0 (en)
MA (1) MA31183B1 (en)
MX (1) MX2009007661A (en)
TN (1) TN2009000291A1 (en)
WO (1) WO2008089459A1 (en)

Families Citing this family (51)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP4834553B2 (en) 2004-09-17 2011-12-14 エーザイ・アール・アンド・ディー・マネジメント株式会社 Pharmaceutical composition
US7429667B2 (en) 2005-01-20 2008-09-30 Ardea Biosciences, Inc. Phenylamino isothiazole carboxamidines as MEK inhibitors
US9095581B2 (en) 2005-07-21 2015-08-04 Ardea Biosciences, Inc. Combinations of MEK inhibitors and Raf kinase inhibitors and uses thereof
JP4989476B2 (en) 2005-08-02 2012-08-01 エーザイ・アール・アンド・ディー・マネジメント株式会社 Methods for assaying the effects of angiogenesis inhibitors
RU2448708C3 (en) 2006-05-18 2017-09-28 Эйсай Ар Энд Ди Менеджмент Ко., Лтд. ANTI-TUMOR MEANS AGAINST THYROID CANCER CANCER
US8227603B2 (en) 2006-08-01 2012-07-24 Cytokinetics, Inc. Modulating skeletal muscle
PT2583970E (en) 2006-08-02 2016-02-08 Cytokinetics Inc Certain chemical entities, compositions and methods comprising imidazopyrimidines
US8299248B2 (en) 2006-08-02 2012-10-30 Cytokinetics, Incorporated Certain 1H-imidazo[4,5-b]pyrazin-2(3H)-ones and 1H-imidazo[4,5-b]pyrazin-2-ols and methods for their use
EP2139478A4 (en) * 2007-03-30 2010-05-05 Cytokinetics Inc Certain chemical entities, compositions and methods
CA2924436A1 (en) * 2007-07-30 2009-02-05 Ardea Biosciences, Inc. Pharmaceutical combinations of n-(3,4-difluoro-2-(2-fluoro-4-iodophenylamino)-6-methoxyphenyl)-1-(2,3-dihydroxypropyl)cyclopropane-1-sulfonamide as inhibitors of mek and methods of use
US8044240B2 (en) 2008-03-06 2011-10-25 Ardea Biosciences Inc. Polymorphic form of N-(S)-(3,4-difluoro-2-(2-fluoro-4-iodophenylamino)-6-methoxyphenyl)-1-(2,3-dihydroxypropyl)cyclopropane-1-sulfonamide and uses thereof
AR071587A1 (en) * 2008-04-21 2010-06-30 Novartis Ag HETEROCICLICAL COMPOUNDS AS MEK INHIBITORS
US10253020B2 (en) 2009-06-12 2019-04-09 Abivax Compounds for preventing, inhibiting, or treating cancer, AIDS and/or premature aging
WO2010143169A2 (en) 2009-06-12 2010-12-16 Société Splicos Compounds useful for treating aids
WO2011047055A2 (en) 2009-10-13 2011-04-21 Allostem Therapeutics Llc Novel mek inhibitors, useful in the treatment of diseases
NZ599132A (en) 2009-10-21 2014-05-30 Boehringer Ingelheim Int Indazole and pyrazolopyridine compounds as ccr1 receptor antagonists
EP2493875B1 (en) 2009-10-27 2014-08-06 Boehringer Ingelheim International GmbH Heterocyclic compounds as ccr1 receptor antagonists
EA201200651A1 (en) * 2009-11-04 2012-12-28 Новартис Аг HETEROCYCLIC SULPHONAMIDE DERIVATIVES APPLICABLE AS MEK INHIBITORS
AU2012265844A1 (en) * 2009-12-08 2013-05-02 Novartis Ag Heterocyclic sulfonamide derivatives
WO2011070030A1 (en) * 2009-12-08 2011-06-16 Novartis Ag Heterocyclic sulfonamide derivatives
EP2563787B1 (en) 2010-04-30 2014-11-26 Boehringer Ingelheim International GmbH Azaindazole amide compounds as ccr1 receptor antagonists
CA2828946C (en) 2011-04-18 2016-06-21 Eisai R&D Management Co., Ltd. Therapeutic agent for tumor
ES2841809T3 (en) 2011-06-03 2021-07-09 Eisai R&D Man Co Ltd Biomarkers to predict and evaluate the degree of response of subjects with thyroid and kidney cancer to lenvatinib compounds
CN103748085A (en) 2011-06-09 2014-04-23 诺华股份有限公司 Heterocyclic sulfonamide derivatives
EA201791043A1 (en) 2011-07-13 2017-09-29 Сайтокинетикс, Инк. COMBINED THERAPY OF SIDE AMIOTROPHIC SCLEROSIS
CN103204822B (en) 2012-01-17 2014-12-03 上海科州药物研发有限公司 Benzoxazole compounds as protein kinase inhibitors, and preparation method and application thereof
AU2013337702A1 (en) 2012-11-02 2015-05-21 Merck Patent Gmbh Method of reducing adverse effects in a cancer patient undergoing treatment with a MEK inhibitor
MX2015004979A (en) 2012-12-21 2015-07-17 Eisai R&D Man Co Ltd Amorphous form of quinoline derivative, and method for producing same.
EP2757161A1 (en) 2013-01-17 2014-07-23 Splicos miRNA-124 as a biomarker of viral infection
AU2014266223B2 (en) 2013-05-14 2020-06-25 Eisai R&D Management Co., Ltd. Biomarkers for predicting and assessing responsiveness of endometrial cancer subjects to lenvatinib compounds
US9827237B2 (en) 2013-07-05 2017-11-28 Abivax Compounds useful for treating diseases caused by retroviruses
CN103739550B (en) * 2014-01-02 2016-06-01 中国药科大学 2,3-dimethyl-6-urea-2H-indazole compounds and its preparation method and application
EP3094736A4 (en) 2014-01-14 2017-10-25 Dana-Farber Cancer Institute, Inc. Compositions and methods for identification, assessment, prevention, and treatment of melanoma using pd-l1 isoforms
EP2974729A1 (en) 2014-07-17 2016-01-20 Abivax Quinoline derivatives for use in the treatment of inflammatory diseases
ES2926687T3 (en) 2014-08-28 2022-10-27 Eisai R&D Man Co Ltd Highly pure quinoline derivative and method for its production
CN105384754B (en) * 2014-09-02 2018-04-20 上海科州药物研发有限公司 Heterocycle compound as kinases inhibitor and its preparation method and application
EP3204516B1 (en) 2014-10-06 2023-04-26 Dana-Farber Cancer Institute, Inc. Angiopoietin-2 biomarkers predictive of anti-immune checkpoint response
HUE064614T2 (en) 2015-02-25 2024-04-28 Eisai R&D Man Co Ltd Method for suppressing bitterness of quinoline derivative
KR102662228B1 (en) 2015-03-04 2024-05-02 머크 샤프 앤드 돔 코포레이션 Combination of PD-1 antagonists and VEGFR/FGFR/RET tyrosine kinase inhibitors to treat cancer
MA41866A (en) 2015-03-31 2018-02-06 Massachusetts Gen Hospital SELF-ASSEMBLING MOLECULES FOR TARGETED DRUG DELIVERY
AU2016279474B2 (en) 2015-06-16 2021-09-09 Eisai R&D Management Co., Ltd. Anticancer agent
WO2017033113A1 (en) 2015-08-21 2017-03-02 Acerta Pharma B.V. Therapeutic combinations of a mek inhibitor and a btk inhibitor
CN105085427B (en) * 2015-08-21 2018-06-05 中国科学院广州生物医药与健康研究院 A kind of benzo [d] isoxazole class compound and its application
US10899745B2 (en) * 2017-03-30 2021-01-26 Bristol-Myers Squibb Company Process for the preparation of 6-(cyclopropaneamido)-4-((2-methoxy-3-(1-methyl-1H-1,2,4-triazol-3-yl)phenyl)amino)-N-(methyl-d3)pyridazine-3-carboxamide
CN113490668A (en) 2018-10-05 2021-10-08 安娜普尔纳生物股份有限公司 Compounds and compositions for treating diseases associated with APJ receptor activity
EP3669873A1 (en) 2018-12-20 2020-06-24 Abivax Quinoline derivatives for use ine the traeatment of inflammation diseases
EA202192575A1 (en) 2019-03-21 2022-01-14 Онксео DBAIT COMPOUNDS IN COMBINATION WITH KINASE INHIBITORS FOR CANCER TREATMENT
CN110229173A (en) * 2019-07-17 2019-09-13 泰州职业技术学院 A kind of 5-6-5 aza-tricycle compound and preparation method thereof
US20220401436A1 (en) 2019-11-08 2022-12-22 INSERM (Institute National de la Santé et de la Recherche Médicale) Methods for the treatment of cancers that have acquired resistance to kinase inhibitors
WO2021148581A1 (en) 2020-01-22 2021-07-29 Onxeo Novel dbait molecule and its use
CN112759552A (en) * 2020-12-31 2021-05-07 武汉九州钰民医药科技有限公司 Synthesis method of semetinib

Family Cites Families (122)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US622068A (en) * 1899-03-28 payne
US1468428A (en) * 1921-07-18 1923-09-18 Amos S Wells Dental molding apparatus
US1896123A (en) * 1925-07-29 1933-02-07 Schweitzer Heinrich Wax dental form and method of making same
US1864365A (en) * 1929-09-06 1932-06-21 Ac Spark Plug Co Process and apparatus for forming ceramic bodies
US2271454A (en) * 1938-06-25 1942-01-27 Dental Res Corp Method of forming a reproducting of an article
US2310448A (en) * 1940-03-11 1943-02-09 Henry H Leib Dental apparatus
US2480048A (en) * 1944-07-10 1949-08-23 William S Rice Casting process
US2474676A (en) * 1946-02-27 1949-06-28 Myerson Tooth Corp Method of forming artificial teeth
US2551812A (en) * 1947-07-14 1951-05-08 Alex A Nelson Process of preparing an artificial denture
US2678470A (en) * 1949-11-25 1954-05-18 H D Justi & Son Inc Polymerizing method
US3390458A (en) * 1965-05-10 1968-07-02 Joseph M. Lytton Method of preparing for dental impressions
US3565387A (en) * 1968-09-17 1971-02-23 Dental Innovations Inc Prefabricated dental pattern having adjusting slot means
US3585723A (en) * 1969-06-20 1971-06-22 Ion Co The Dental crown and method of installation thereof
GB1408265A (en) * 1971-10-18 1975-10-01 Ici Ltd Photopolymerisable composition
US3949476A (en) * 1974-03-25 1976-04-13 Henry Kahn Device useful in dental crown procedures and method of using the same
US4024636A (en) * 1975-07-11 1977-05-24 Polythetics, Inc. Dentures
US4024637A (en) * 1975-07-11 1977-05-24 Polythetics, Inc. Dentures and process for making the same
US4113499A (en) * 1976-03-18 1978-09-12 Valentin Nikolaevich Ivanov Suspension for making molds in disposable pattern casting
DE2759214A1 (en) * 1977-12-31 1979-07-05 Scheicher Hans METHOD OF MANUFACTURING IMPLANTS
DE3071095D1 (en) * 1979-07-13 1985-10-24 Corning Glass Works Dental constructs and tools and production thereof
US4585417A (en) * 1979-12-14 1986-04-29 Coors Porcelain Company Dental appliance and method of manufacture
US4347888A (en) * 1980-03-20 1982-09-07 Butler Melvyn P Method of making forms for investment casting and products produced therefrom
DE3110009A1 (en) * 1981-03-11 1982-09-30 Schering Ag, 1000 Berlin Und 4619 Bergkamen NEW BENZOFURAN DERIVATIVES, THEIR PRODUCTION AND USE
DE3135469C1 (en) * 1981-09-08 1983-04-21 Peter 6272 Niedernhausen Bayer Process for the production of dentures, dental prostheses or lost casting models for this and molds for carrying out this process
US4514174A (en) * 1982-11-19 1985-04-30 Dentsply Research & Development Corp. Methods for posterior dental restoration employing light curable packable compositions
US4568737A (en) * 1983-01-07 1986-02-04 The Dow Chemical Company Dense star polymers and dendrimers
US4737550A (en) * 1983-01-07 1988-04-12 The Dow Chemical Company Bridged dense star polymers
CH662266A5 (en) * 1983-10-04 1987-09-30 Weissenfluh Hans Dr Von FILM-SHAPED DENTAL MATRICE.
US4503169A (en) * 1984-04-19 1985-03-05 Minnesota Mining And Manufacturing Company Radiopaque, low visual opacity dental composites containing non-vitreous microparticles
JPS60252083A (en) * 1984-05-28 1985-12-12 本田技研工業株式会社 Air intake device in car
US4571188A (en) * 1984-07-05 1986-02-18 Sybron Corporation Occlusal matrix for light cured composites
US4587329A (en) * 1984-08-17 1986-05-06 The Dow Chemical Company Dense star polymers having two dimensional molecular diameter
US4642126A (en) * 1985-02-11 1987-02-10 Norton Company Coated abrasives with rapidly curable adhesives and controllable curvature
US4648843A (en) * 1985-07-19 1987-03-10 Minnesota Mining And Manufacturing Company Method of dental treatment using poly(ethylenically unsaturated) carbamoyl isocyanurates and dental materials made therewith
US4652274A (en) * 1985-08-07 1987-03-24 Minnesota Mining And Manufacturing Company Coated abrasive product having radiation curable binder
US4694064A (en) * 1986-02-28 1987-09-15 The Dow Chemical Company Rod-shaped dendrimer
CA1323949C (en) * 1987-04-02 1993-11-02 Michael C. Palazzotto Ternary photoinitiator system for addition polymerization
US4767331A (en) * 1987-05-28 1988-08-30 Hoe Khin A Dental crown manufacturing apparatus
US4857599A (en) * 1988-02-08 1989-08-15 The Dow Chemical Company Modified dense star polymers
GB8827305D0 (en) 1988-11-23 1988-12-29 British Bio Technology Compounds
US5348154A (en) * 1989-05-10 1994-09-20 Minnesota Mining And Manufacturing Company Packaging curable materials
DK0443269T3 (en) * 1990-02-23 1993-12-27 Minnesota Mining & Mfg Semi-thermoplastic molding material with heat-stable memory of individually adapted shape
US5066231A (en) * 1990-02-23 1991-11-19 Minnesota Mining And Manufacturing Company Dental impression process using polycaprolactone molding composition
US5102332A (en) * 1991-02-25 1992-04-07 Ticore Dental Systems Braided fiber dental retainer and container therefor
US5135545A (en) * 1991-03-22 1992-08-04 The Dow Chemical Company Method for making machinable abrasive greenware
US5192207A (en) * 1991-09-17 1993-03-09 Rosellini Davey G Composite resin crown, replacement tooth and method
SE9200564L (en) * 1992-02-26 1993-03-15 Perstorp Ab DENDRITIC MACROMOLECYLE OF POLYESTER TYPE, PROCEDURES FOR PRODUCING THEREOF AND USING THEREOF
US5455258A (en) 1993-01-06 1995-10-03 Ciba-Geigy Corporation Arylsulfonamido-substituted hydroxamic acids
US5914185A (en) * 1993-01-19 1999-06-22 Shoher; Itzhak Moldable dental material composition
US5401169A (en) * 1993-06-10 1995-03-28 Minnesota Mining And Manufacturing Multiple-part dental material delivery system
JP3690825B2 (en) * 1993-07-26 2005-08-31 エーザイ株式会社 Tricyclic heterocycle-containing sulfonamides and sulfonic acid ester derivatives
US5834462A (en) * 1993-07-26 1998-11-10 Eisai Co., Ltd. Tricyclic heterocyclic sulfonamide and sulfonic ester derivatives
US5487663A (en) * 1993-08-16 1996-01-30 Wilson; George M. Oral appliances and method
US5863949A (en) 1995-03-08 1999-01-26 Pfizer Inc Arylsulfonylamino hydroxamic acid derivatives
US5538129A (en) * 1995-03-21 1996-07-23 Minnesota Mining And Manufacturing Company Package for adhesive precoated dental appliance
JP3053222B2 (en) 1995-04-20 2000-06-19 ファイザー・インコーポレーテッド Arylsulfonylhydroxamic acid derivatives as MMP and TNF inhibitors
HU225506B1 (en) * 1995-07-19 2007-01-29 Merck & Co Inc Use of 4-(4-methylsulfonyl-phenyl)-3-phenyl-2-(5h)-furanone for the preparation of medicaments treating colonic adenomas
US5919870A (en) * 1995-07-31 1999-07-06 Fmc Corporation Functional telechelic star polymers
ATE225343T1 (en) 1995-12-20 2002-10-15 Hoffmann La Roche MATRIX METALLOPROTEASE INHIBITORS
US6057383A (en) * 1996-06-18 2000-05-02 Ivoclar Ag Dental material based on polymerizable waxes
EP0818442A3 (en) 1996-07-12 1998-12-30 Pfizer Inc. Cyclic sulphone derivatives as inhibitors of metalloproteinases and of the production of tumour necrosis factor
TR199900066T2 (en) 1996-07-18 1999-04-21 Pfizer Inc. Matrix metalloprotateazlar�n phosphinate bazl� inhibit�rleri
SK21499A3 (en) 1996-08-23 2000-05-16 Pfizer Arylsulfonylamino hydroxamic acid derivatives
US5797236A (en) * 1996-09-09 1998-08-25 Posey, Jr.; John T. Auxiliary bottom insert apparatus for a container
US5785178A (en) * 1996-11-04 1998-07-28 Minnesota Mining And Manufacturing Co. Packaged photocurable composition
US6077864A (en) 1997-01-06 2000-06-20 Pfizer Inc. Cyclic sulfone derivatives
DE69817801T2 (en) 1997-02-03 2004-03-11 Pfizer Products Inc., Groton ARYLSULFONYLHYDROXAMSÄUREDERIVATE
BR9807824A (en) 1997-02-07 2000-03-08 Pfizer Derivatives of n-hydroxy-beta-sulfonyl-propionamide and its use as matrix metalloproteinase inhibitors
EP0960098A1 (en) 1997-02-11 1999-12-01 Pfizer Inc. Arylsulfonyl hydroxamic acid derivatives
US6353040B1 (en) * 1997-04-02 2002-03-05 Dentsply Research & Development Corp. Dental composite restorative material and method of restoring a tooth
JPH10277061A (en) * 1997-04-04 1998-10-20 Injietsukusu:Kk Production of abutment tooth model and crown restoration material
USD403768S (en) * 1997-05-09 1999-01-05 Minnesota Mining And Manufacturing Company Fiber tip applicator
US5775913A (en) * 1997-05-27 1998-07-07 Updyke; John R. Process for minimal time tooth capping
US6506816B1 (en) * 1997-07-17 2003-01-14 3M Innovative Properties Company Dental resin cements having improved handling properties
TR200000368T2 (en) 1997-08-08 2000-07-21 Pfizer Products Inc. Ariloxyariarylsulfonylamino hydroxamic acid derivatives.
DE19736471A1 (en) * 1997-08-21 1999-02-25 Espe Dental Ag Light-induced cationic curing compositions and their use
US6002336A (en) 1997-12-02 1999-12-14 Lynx System Developers, Inc. Reaction time measurement system
GB9725782D0 (en) 1997-12-05 1998-02-04 Pfizer Ltd Therapeutic agents
US6345984B2 (en) * 1998-04-13 2002-02-12 Jeneric/Pentron, Inc. Prefabricated components for dental appliances
US6186790B1 (en) * 1998-04-13 2001-02-13 Jeneric/Pentron Incorporated Prefabricated components for dental appliances
GB9801690D0 (en) 1998-01-27 1998-03-25 Pfizer Ltd Therapeutic agents
US5876209A (en) * 1998-02-27 1999-03-02 Letcher; William F. Method of manufacturing a dental crown
JP4462654B2 (en) 1998-03-26 2010-05-12 ソニー株式会社 Video material selection device and video material selection method
PA8469501A1 (en) 1998-04-10 2000-09-29 Pfizer Prod Inc HYDROXAMIDES OF THE ACID (4-ARILSULFONILAMINO) -TETRAHIDROPIRAN-4-CARBOXILICO
PA8469401A1 (en) 1998-04-10 2000-05-24 Pfizer Prod Inc BICYCLE DERIVATIVES OF HYDROXAMIC ACID
US6187836B1 (en) * 1998-06-05 2001-02-13 3M Innovative Properties Company Compositions featuring cationically active and free radically active functional groups, and methods for polymerizing such compositions
US6359090B1 (en) * 1998-06-09 2002-03-19 Kerr Corporation Polymerizable dispersant
US6127450A (en) * 1998-06-09 2000-10-03 Kerr Corporation Dental restorative composite
IT245694Y1 (en) * 1998-07-27 2002-03-22 Rini Guido PACKAGING FOR ARTIFICIAL TEETH FOR PROSTHETIC USE
US6252014B1 (en) * 1998-08-04 2001-06-26 Colorado School Of Mines Star polymers and polymeric particles in the nanometer-sized range by step growth reactions
US6196840B1 (en) * 1998-08-14 2001-03-06 Dentsply Research & Development Corp. Custom fitting variable dimension dental impression tray, product and method
DE69915004T2 (en) 1998-11-05 2004-09-09 Pfizer Products Inc., Groton 5-Oxo-pyrrolidine-2-carboxylic acid Hydroxamidderivate
CA2349832A1 (en) * 1999-01-13 2000-07-20 Warner-Lambert Company Benzenesulfonamide derivatives and their use as mek inhibitors
US6318994B1 (en) * 1999-05-13 2001-11-20 Align Technology, Inc Tooth path treatment plan
GB9912961D0 (en) 1999-06-03 1999-08-04 Pfizer Ltd Metalloprotease inhibitors
AU5785900A (en) * 1999-07-16 2001-02-05 Warner-Lambert Company Method for treating chronic pain using mek inhibitors
US6183249B1 (en) * 1999-07-29 2001-02-06 3M Innovative Properties Company Release substrate for adhesive precoated orthodontic appliances
US6572693B1 (en) * 1999-10-28 2003-06-03 3M Innovative Properties Company Aesthetic dental materials
US6592369B2 (en) * 2000-09-26 2003-07-15 Dentsply Research & Development Corp. Wax-like polymerizable dental material, method and shaped product
US7566412B2 (en) * 1999-11-10 2009-07-28 Dentsply International Inc. Dental method and device
US7175433B2 (en) * 1999-11-10 2007-02-13 Dentsply International Inc. Dental material and method
US20020117393A1 (en) * 2000-10-13 2002-08-29 Sun Benjamin J. Licht curing system and method
US6382980B1 (en) * 2000-03-21 2002-05-07 Itzhak Shoher Compact dental multi-layered material for crown and bridge prosthodontics and method
WO2001074301A1 (en) * 2000-04-03 2001-10-11 3M Innovative Properties Company Dental materials with extendable work time, kits, and methods
US6448301B1 (en) * 2000-09-08 2002-09-10 3M Innovative Properties Company Crosslinkable polymeric compositions and use thereof
US20070018346A1 (en) * 2000-09-26 2007-01-25 Sun Benjamin J Light curing system and method
US20020102519A1 (en) * 2001-01-26 2002-08-01 Lloyd Baum Dental prostheses fabrication method using pre-contoured impressionable pattern
US6635690B2 (en) * 2001-06-19 2003-10-21 3M Innovative Properties Company Reactive oligomers
CA2454617A1 (en) * 2001-08-15 2003-02-27 3M Innovative Properties Company Hardenable self-supporting structures and methods
US7134875B2 (en) * 2002-06-28 2006-11-14 3M Innovative Properties Company Processes for forming dental materials and device
US8956160B2 (en) * 2002-07-02 2015-02-17 Ranir, Llc Device and method for delivering an oral care agent
CN100579579C (en) 2002-10-01 2010-01-13 诺华疫苗和诊断公司 Anti-cancer and anti-infectious disease compositions and methods for using same
US6989124B2 (en) * 2003-01-24 2006-01-24 Husky Injection Molding Systems Ltd. Apparatus and method for removing a molded article from a mold
TW200505834A (en) * 2003-03-18 2005-02-16 Sankyo Co Sulfamide derivative and the pharmaceutical composition thereof
US20050042576A1 (en) * 2003-08-19 2005-02-24 Oxman Joel D. Dental article forms and methods
US20050042577A1 (en) * 2003-08-19 2005-02-24 Kvitrud James R. Dental crown forms and methods
US20050040551A1 (en) * 2003-08-19 2005-02-24 Biegler Robert M. Hardenable dental article and method of manufacturing the same
DK1689233T3 (en) 2003-11-19 2012-10-15 Array Biopharma Inc Bicyclic inhibitors of MEK
US20050147944A1 (en) * 2003-12-31 2005-07-07 Naimul Karim Curable dental mill blanks and related methods
DE602005016718D1 (en) * 2004-12-01 2009-10-29 Merck Serono Sa Coinsins Ä1,2,4ÜTRIAZOLOÄ4,3-AÜPYRIDIN DERIVATIVE FOR THE TREATMENT OF HYPERPROLIFERATIVE DISEASES
PL1912636T3 (en) * 2005-07-21 2015-02-27 Ardea Biosciences Inc N-(arylamino)-sulfonamide inhibitors of mek
US7960567B2 (en) * 2007-05-02 2011-06-14 Amgen Inc. Compounds and methods useful for treating asthma and allergic inflammation

Also Published As

Publication number Publication date
US20080255133A1 (en) 2008-10-16
ES2547303T3 (en) 2015-10-05
EA200900959A1 (en) 2010-02-26
CA2675358C (en) 2016-01-05
CR10939A (en) 2009-11-13
CN101663279A (en) 2010-03-03
DOP2009000181A (en) 2010-04-30
US8063049B2 (en) 2011-11-22
AU2008206045A1 (en) 2008-07-24
CA2675358A1 (en) 2008-07-24
MA31183B1 (en) 2010-02-01
MX2009007661A (en) 2009-12-14
KR20090111847A (en) 2009-10-27
TN2009000291A1 (en) 2010-12-31
US20100331334A1 (en) 2010-12-30
EP2121620A1 (en) 2009-11-25
IL199727A0 (en) 2010-04-15
WO2008089459A1 (en) 2008-07-24
ECSP099525A (en) 2009-11-30
JP5491199B2 (en) 2014-05-14
CO6210813A2 (en) 2010-10-20
JP2010516698A (en) 2010-05-20
US7820664B2 (en) 2010-10-26
BRPI0806898A2 (en) 2015-07-14

Similar Documents

Publication Publication Date Title
EP2121620B1 (en) Inhibitors of mek
US11827644B2 (en) Pyrazine derivative and application thereof in inhibiting SHP2
US7144907B2 (en) Heterocyclic inhibitors of MEK and methods of use thereof
BR112019015720A2 (en) COMPOUND, PHARMACEUTICAL COMPOSITION, AND, USE OF A COMPOUND OR PHARMACEUTICAL COMPOSITION
WO2012159565A1 (en) 6-(aryl-carboxamide) imidazo [1,2-a] pyrimidine and 6-(aryl carboxamide) [1,2,4] triazolo [4,3-a] pyrimidine as hedgehog pathway inhibitor and use thereof
AU2018226922B2 (en) Urea-substituted aromatic ring-linked dioxane-quinazoline and -linked dioxane-quinoline compounds, preparation method therefor and use thereof
CZ411097A3 (en) Aromatic compounds, processes of their preparation and pharmaceutical preparation in which they are comprised
BR112018016708B1 (en) 7H-PYRROLO[2,3-D]PYRIMIDINE COMPOUNDS AND PROCESS FOR PREPARING SAID COMPOUNDS
CA2892304C (en) Compounds as diacylglycerol acyltransferase inhibitors
CN117069696A (en) Double-target small molecule inhibitor and preparation method and application thereof

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20090819

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MT NL NO PL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: AL BA MK RS

17Q First examination report despatched

Effective date: 20111122

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

INTG Intention to grant announced

Effective date: 20150226

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MT NL NO PL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: AL BA MK RS

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

RAP2 Party data changed (patent owner data changed or rights of a patent transferred)

Owner name: ARDEA BIOSCIENCES, INC.

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: AT

Ref legal event code: REF

Ref document number: 731838

Country of ref document: AT

Kind code of ref document: T

Effective date: 20150715

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602008038605

Country of ref document: DE

REG Reference to a national code

Ref country code: ES

Ref legal event code: FG2A

Ref document number: 2547303

Country of ref document: ES

Kind code of ref document: T3

Effective date: 20151005

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: NO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150917

Ref country code: HR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150617

Ref country code: FI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150617

Ref country code: LT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150617

REG Reference to a national code

Ref country code: AT

Ref legal event code: MK05

Ref document number: 731838

Country of ref document: AT

Kind code of ref document: T

Effective date: 20150617

REG Reference to a national code

Ref country code: LT

Ref legal event code: MG4D

Ref country code: NL

Ref legal event code: MP

Effective date: 20150617

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150918

Ref country code: BG

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150917

Ref country code: LV

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150617

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 9

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: EE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150617

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20151019

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150617

Ref country code: CZ

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150617

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20151017

Ref country code: RO

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20150617

Ref country code: PL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150617

Ref country code: AT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150617

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602008038605

Country of ref document: DE

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: DK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150617

26N No opposition filed

Effective date: 20160318

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20160131

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150617

Ref country code: LU

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20160118

REG Reference to a national code

Ref country code: CH

Ref legal event code: PL

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MC

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150617

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CH

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20160131

Ref country code: LI

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20160131

REG Reference to a national code

Ref country code: IE

Ref legal event code: MM4A

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150617

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 10

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20160118

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150617

Ref country code: NL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150617

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150617

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 11

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: DE

Payment date: 20180129

Year of fee payment: 11

Ref country code: ES

Payment date: 20180201

Year of fee payment: 11

Ref country code: GB

Payment date: 20180129

Year of fee payment: 11

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: HU

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT; INVALID AB INITIO

Effective date: 20080118

Ref country code: CY

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150617

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: FR

Payment date: 20180125

Year of fee payment: 11

Ref country code: IT

Payment date: 20180122

Year of fee payment: 11

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20160131

Ref country code: TR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150617

REG Reference to a national code

Ref country code: DE

Ref legal event code: R119

Ref document number: 602008038605

Country of ref document: DE

GBPC Gb: european patent ceased through non-payment of renewal fee

Effective date: 20190118

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: DE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20190801

Ref country code: FR

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20190131

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: GB

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20190118

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20190118

REG Reference to a national code

Ref country code: ES

Ref legal event code: FD2A

Effective date: 20200310

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: ES

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20190119